1
|
Morales ED, Wang D, Burke MJ, Han J, Devine DD, Zhang K, Duan D. Transcriptional changes of genes encoding sarcoplasmic reticulum calcium binding and up-taking proteins in normal and Duchenne muscular dystrophy dogs. BMC Musculoskelet Disord 2024; 25:811. [PMID: 39402529 PMCID: PMC11472500 DOI: 10.1186/s12891-024-07927-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Cytosolic calcium overload contributes to muscle degradation in Duchenne muscular dystrophy (DMD). The sarcoplasmic reticulum (SR) is the primary calcium storage organelle in muscle. The sarco-endoplasmic reticulum ATPase (SERCA) pumps cytosolic calcium to the SR during muscle relaxation. Calcium is kept in the SR by calcium-binding proteins. METHODS Given the importance of the canine DMD model in translational studies, we examined transcriptional changes of SERCA (SERCA1 and SERCA2a), SERCA regulators (phospholamban, sarcolipin, myoregulin, and dwarf open reading frame), and SR calcium-binding proteins (calreticulin, calsequestrin 1, calsequestrin 2, and sarcalumenin) in skeletal muscle (diaphragm and extensor carpi ulnaris) and heart (left ventricle) in normal and affected male dogs by droplet digital PCR before the onset (≤ 2-m-old), at the active stage (8 to 16-m-old), and at the terminal stage (30 to 50-m-old) of the disease. Since many of these proteins are expressed in a fiber type-specific manner, we also evaluated fiber type composition in skeletal muscle. RESULTS In affected dog skeletal muscle, SERCA and its regulators were down-regulated at the active stage, but calcium-binding proteins (except for calsequestrin 1) were upregulated at the terminal stage. Surprisingly, nominal differences were detected in the heart. We also examined whether there exists sex-biased expression in 8 to 16-m-old dogs. Multiple transcripts were significantly downregulated in the heart and extensor carpi ulnaris muscle of female dogs. In fiber type analysis, we found significantly more type I fiber in the diaphragm of 8 to 16-m-old affected dogs, and significantly more type II fibers in the extensor carpi ulnaris of 30 to 50-m-old affected dogs. However, no difference was detected between male and female dogs. CONCLUSIONS Our study adds new knowledge to the understanding of muscle calcium regulation in normal and dystrophic canines.
Collapse
Affiliation(s)
- Emily D Morales
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Dongxin Wang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Matthew J Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Jin Han
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Drake D Devine
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO, 65212, USA.
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
2
|
Paula S, Floruta S, Pajazetovic K, Sobota S, Almahmodi D. The molecular determinants of calcium ATPase inhibition by curcuminoids. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184367. [PMID: 38969202 DOI: 10.1016/j.bbamem.2024.184367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
The natural product curcumin and some of its analogs are known inhibitors of the transmembrane enzyme sarco/endoplasmic reticulum calcium ATPase (SERCA). Despite their widespread use, the curcuminoids' binding site in SERCA and their relevant interactions with the enzyme remain elusive. This lack of knowledge has prevented the development of curcuminoids into valuable experimental tools or into agents of therapeutic value. We used the crystal structures of SERCA in its E1 conformation in conjunction with computational tools such as docking and surface screens to determine the most likely curcumin binding site, along with key enzyme/inhibitor interactions. Additionally, we determined the inhibitory potencies and binding affinities for a small set of curcumin analogs. The predicted curcumin binding site is a narrow cleft in the transmembrane section of SERCA, close to the transmembrane/cytosol interface. In addition to pronounced complementarity in shape and hydrophobicity profiles between curcumin and the binding pocket, several hydrogen bonds were observed that were spread over the entire curcumin scaffold, involving residues on several transmembrane helices. Docking-predicted interactions were compatible with experimental observations for inhibitory potencies and binding affinities. Based on these findings, we propose an inhibition mechanism that assumes that the presence of a curcuminoid in the binding site arrests the catalytic cycle of SERCA by preventing it from converting from the E1 to the E2 conformation. This blockage of conformational change is accomplished by a combination of steric hinderance and hydrogen-bond-based cross-linking of transmembrane helices that require flexibility throughout the catalytic cycle.
Collapse
Affiliation(s)
- Stefan Paula
- Department of Chemistry, California State University Sacramento, 6000 J Street, Sacramento, CA 95819, USA.
| | - Sergiu Floruta
- Department of Chemistry, California State University Sacramento, 6000 J Street, Sacramento, CA 95819, USA
| | - Karim Pajazetovic
- Department of Chemistry, California State University Sacramento, 6000 J Street, Sacramento, CA 95819, USA
| | - Sydni Sobota
- Department of Chemistry, California State University Sacramento, 6000 J Street, Sacramento, CA 95819, USA
| | - Dina Almahmodi
- Department of Chemistry, California State University Sacramento, 6000 J Street, Sacramento, CA 95819, USA
| |
Collapse
|
3
|
Shapiro IM, Risbud MV, Landis WJ. Toward understanding the cellular control of vertebrate mineralization: The potential role of mitochondria. Bone 2024; 185:117112. [PMID: 38697384 PMCID: PMC11251007 DOI: 10.1016/j.bone.2024.117112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/17/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
This review examines the possible role of mitochondria in maintaining calcium and phosphate ion homeostasis and participating in the mineralization of bone, cartilage and other vertebrate hard tissues. The paper builds on the known structural features of mitochondria and the documented observations in these tissues that the organelles contain calcium phosphate granules. Such deposits in mitochondria putatively form to buffer excessively high cytosolic calcium ion concentrations and prevent metabolic deficits and even cell death. While mitochondria protect cytosolic enzyme systems through this buffering capacity, the accumulation of calcium ions by mitochondria promotes the activity of enzymes of the tricarboxylic acid (TCA/Krebs) cycle, increases oxidative phosphorylation and ATP synthesis, and leads to changes in intramitochondrial pH. These pH alterations influence ion solubility and possibly the transitions and composition in the mineral phase structure of the granules. Based on these considerations, mitochondria are proposed to support the mineralization process by providing a mobile store of calcium and phosphate ions, in smaller cluster or larger granule form, while maintaining critical cellular activities. The rise in the mitochondrial calcium level also increases the generation of citrate and other TCA cycle intermediates that contribute to cell function and the development of extracellular mineral. This paper suggests that another key role of the mitochondrion, along with the effects just noted, is to supply phosphate ions, derived from the breakdown of ATP, to endolysosomes and autophagic vesicles originating in the endoplasmic reticulum and Golgi and at the plasma membrane. These many separate but interdependent mitochondrial functions emphasize the critical importance of this organelle in the cellular control of vertebrate mineralization.
Collapse
Affiliation(s)
- Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America.
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - William J Landis
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California at San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
4
|
Peng M, Zhou Y, Wan C. Identification of phosphorylated small ORF-encoded peptides in Hep3B cells by LC/MS/MS. J Proteomics 2024; 303:105214. [PMID: 38823442 DOI: 10.1016/j.jprot.2024.105214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Small ORF-encoded peptides (SEPs) are a class of low molecular weight proteins and peptides comprising <100 amino acids with important functions in various life activities. Although the sequence length is short, SEPs might also have post-translational modification (PTM). Phosphorylation is one of the most essential PTMs of proteins. In this work, we enriched phosphopeptides with IMAC and TiO2 materials and analyzed the phosphorylated SEPs in Hep3B cells. A total of 24 phosphorylated SEPs were identified, and 11 SEPs were coded by ncRNA. For the sequence analysis, we found that the general characteristics of phosphorylated SEPs are roughly the same as canonical proteins. Besides, two phosphorylation SEPs have the Stathmin family signature 2 motif, which can regulate the microtubule cytoskeleton. Some SEPs have domains or signal peptides, indicating their specific functions and subcellular locations. Kinase network analysis found a small number of kinases that may be a clue to the specific functions of some SEPs. However, only one-fifth of the predicted phosphorylation sites were identified by LC/MS/MS, indicating that many SEP PTMs are hidden in the dark, waiting to be uncovered and verified. This study helps expand our understanding of SEP and provides information for further SEP function investigation. SIGNIFICANCE: Small ORF-encoded peptides (SEPs) are important in various life activities. Although the sequence length is short (<100AA), SEPs might also have post-translational modification (PTM). Phosphorylation is one of the most essential PTMs of proteins. We enriched phosphopeptides and analyzed the phosphorylated SEPs in Hep3B cells. That is the first time to explore the PTM of SPEs systematically. Kinase network analysis found a small number of kinases that may be a clue to the specific functions of SEPs. More SEP PTMs are hidden in the dark and waiting to be uncovered and verified. This study helps expand our understanding of SEP and provides information for further SEP function investigation.
Collapse
Affiliation(s)
- Mingbo Peng
- School of Life Sciences, and Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, Hubei 430079, People's Republic of China
| | - Yutian Zhou
- School of Life Sciences, and Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, Hubei 430079, People's Republic of China
| | - Cuihong Wan
- School of Life Sciences, and Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, Hubei 430079, People's Republic of China.
| |
Collapse
|
5
|
Bovo E, Jamrozik T, Kahn D, Karkut P, Robia SL, Zima AV. Phosphorylation of phospholamban promotes SERCA2a activation by dwarf open reading frame (DWORF). Cell Calcium 2024; 121:102910. [PMID: 38823350 PMCID: PMC11247691 DOI: 10.1016/j.ceca.2024.102910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024]
Abstract
In cardiac myocytes, the type 2a sarco/endoplasmic reticulum Ca-ATPase (SERCA2a) plays a key role in intracellular Ca regulation. Due to its critical role in heart function, SERCA2a activity is tightly regulated by different mechanisms, including micropeptides. While phospholamban (PLB) is a well-known SERCA2a inhibitor, dwarf open reading frame (DWORF) is a recently identified SERCA2a activator. Since PLB phosphorylation is the most recognized mechanism of SERCA2a activation during adrenergic stress, we studied whether PLB phosphorylation also affects SERCA2a regulation by DWORF. By using confocal Ca imaging in a HEK293 expressing cell system, we analyzed the effect of the co-expression of PLB and DWORF using a bicistronic construct on SERCA2a-mediated Ca uptake. Under these conditions of matched expression of PLB and DWORF, we found that SERCA2a inhibition by non-phosphorylated PLB prevails over DWORF activating effect. However, when PLB is phosphorylated at PKA and CaMKII sites, not only PLB's inhibitory effect was relieved, but SERCA2a was effectively activated by DWORF. Förster resonance energy transfer (FRET) analysis between SERCA2a and DWORF showed that DWORF has a higher relative affinity for SERCA2a when PLB is phosphorylated. Thus, SERCA2a regulation by DWORF responds to the PLB phosphorylation status, suggesting that DWORF might contribute to SERCA2a activation during conditions of adrenergic stress.
Collapse
Affiliation(s)
- Elisa Bovo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA.
| | - Thomas Jamrozik
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Daniel Kahn
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Patryk Karkut
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| | - Aleksey V Zima
- Department of Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, 2160 South First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
6
|
Wang L, Zhang J, Li W, Zhang X, Yokoyama T, Sakamoto M, Wang Y. The A-kinase anchoring protein Yotiao decrease the ER calcium content by inhibiting the store operated calcium entry. Cell Calcium 2024; 121:102906. [PMID: 38781694 DOI: 10.1016/j.ceca.2024.102906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024]
Abstract
The meticulous regulation of ER calcium (Ca2+) homeostasis is indispensable for the proper functioning of numerous cellular processes. Disrupted ER Ca2+ balance is implicated in diverse diseases, underscoring the need for a systematic exploration of its regulatory factors in cells. Our recent genomic-scale screen identified a scaffolding protein A-kinase anchoring protein 9 (AKAP9) as a regulator of ER Ca2+ levels, but the underlying molecular mechanisms remain elusive. Here, we reveal that Yotiao, the smallest splicing variant of AKAP9 decreased ER Ca2+ content in animal cells. Additional testing using a combination of Yotiao truncations, knock-out cells and pharmacological tools revealed that, Yotiao does not require most of its interactors, including type 1 inositol 1,4,5-trisphosphate receptors (IP3R1), protein kinase A (PKA), protein phosphatase 1 (PP1), adenylyl cyclase type 2 (AC2) and so on, to reduce ER Ca2+ levels. However, adenylyl cyclase type 9 (AC9), which is known to increases its cAMP generation upon interaction with Yotiao for the modulation of potassium channels, plays an essential role for Yotiao's ER-Ca2+-lowering effect. Mechanistically, Yotiao may work through AC9 to act on Orai1-C terminus and suppress store operated Ca2+ entry, resulting in reduced ER Ca2+ levels. These findings not only enhance our comprehension of the interplay between Yotiao and AC9 but also contribute to a more intricate understanding of the finely tuned mechanisms governing ER Ca2+ homeostasis.
Collapse
Affiliation(s)
- Liuqing Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Jiaxuan Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Wanjie Li
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Xiaoyan Zhang
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Tatsushi Yokoyama
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Masayuki Sakamoto
- Department of Optical Neural and Molecular Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, China; Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
7
|
Chandan K, Gupta M, Ahmad A, Sarwat M. P-type calcium ATPases play important roles in biotic and abiotic stress signaling. PLANTA 2024; 260:37. [PMID: 38922354 DOI: 10.1007/s00425-024-04462-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/09/2024] [Indexed: 06/27/2024]
Abstract
MAIN CONCLUSION Knowledge of Ca2+-ATPases is imperative for improving crop quality/ food security, highly threatened due to global warming. Ca2+-ATPases modulates calcium, essential for stress signaling and modulating growth, development, and immune activities. Calcium is considered a versatile secondary messenger and essential for short- and long-term responses to biotic and abiotic stresses in plants. Coordinated transport activities from both calcium influx and efflux channels are required to generate cellular calcium signals. Various extracellular stimuli cause an induction in cytosolic calcium levels. To cope with such stresses, it is important to maintain intracellular Ca2+ levels. Plants need to evolve efficient efflux mechanisms to maintain Ca2+ ion homeostasis. Plant Ca2+-ATPases are members of the P-type ATPase superfamily and localized in the plasma membrane and endoplasmic reticulum (ER). They are required for various cellular processes, including plant growth, development, calcium signaling, and even retorts to environmental stress. These ATPases play an essential role in Ca2+ homeostasis and are actively involved in Ca2+ transport. Plant Ca2+-ATPases are categorized into two major classes: type IIA and type IIB. Although these two classes of ATPases share similarities in protein sequence, they differ in their structure, cellular localization, and sensitivity to inhibitors. Due to the emerging role of Ca2+-ATPase in abiotic and biotic plant stress, members of this family may help promote agricultural improvement under stress conditions. This review provides a comprehensive overview of P-type Ca2+-ATPase, and their role in Ca2+ transport, stress signaling, and cellular homeostasis focusing on their classification, evolution, ion specificities, and catalytic mechanisms. It also describes the main aspects of the role of Ca2+-ATPase in transducing signals during plant biotic and abiotic stress responses and its role in plant development and physiology.
Collapse
Affiliation(s)
- Kumari Chandan
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, 201313, India
| | - Meenakshi Gupta
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, 201313, India
| | - Altaf Ahmad
- Department of Botany, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, 201313, India.
| |
Collapse
|
8
|
Park M, Mun SY, Zhuang W, Jeong J, Kim HR, Park H, Han ET, Han JH, Chun W, Li H, Park WS. The antidiabetic drug ipragliflozin induces vasorelaxation of rabbit femoral artery by activating a Kv channel, the SERCA pump, and the PKA signaling pathway. Eur J Pharmacol 2024; 972:176589. [PMID: 38631503 DOI: 10.1016/j.ejphar.2024.176589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/29/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
We explored the vasorelaxant effects of ipragliflozin, a sodium-glucose cotransporter-2 inhibitor, on rabbit femoral arterial rings. Ipragliflozin relaxed phenylephrine-induced pre-contracted rings in a dose-dependent manner. Pre-treatment with the ATP-sensitive K+ channel inhibitor glibenclamide (10 μM), the inwardly rectifying K+ channel inhibitor Ba2+ (50 μM), or the Ca2+-sensitive K+ channel inhibitor paxilline (10 μM) did not influence the vasorelaxant effect. However, the voltage-dependent K+ (Kv) channel inhibitor 4-aminopyridine (3 mM) reduced the vasorelaxant effect. Specifically, the vasorelaxant response to ipragliflozin was significantly attenuated by pretreatment with the Kv7.X channel inhibitors linopirdine (10 μM) and XE991 (10 μM), the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin (1 μM) and cyclopiazonic acid (10 μM), and the cAMP/protein kinase A (PKA)-associated signaling pathway inhibitors SQ22536 (50 μM) and KT5720 (1 μM). Neither the cGMP/protein kinase G (PKG)-associated signaling pathway nor the endothelium was involved in ipragliflozin-induced vasorelaxation. We conclude that ipragliflozin induced vasorelaxation of rabbit femoral arteries by activating Kv channels (principally the Kv7.X channel), the SERCA pump, and the cAMP/PKA-associated signaling pathway independent of other K+ (ATP-sensitive K+, inwardly rectifying K+, and Ca2+-sensitive K+) channels, cGMP/PKG-associated signaling, and the endothelium.
Collapse
Affiliation(s)
- Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hye Ryung Kim
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongliang Li
- Institute of Translational Medicine, Medical College, Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment for Senile Diseases, Yangzhou University, Yangzhou, 225001, Jiangsu, China.
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
9
|
Saloman JL, Epouhe AY, Ruff CF, Albers KM. PDX1, a transcription factor essential for organ differentiation, regulates SERCA-dependent Ca 2+ homeostasis in sensory neurons. Cell Calcium 2024; 120:102884. [PMID: 38574509 PMCID: PMC11188734 DOI: 10.1016/j.ceca.2024.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/13/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
Pancreatic and duodenal homeobox 1 (PDX1) is a transcription factor required for the development and differentiation of the pancreas. Previous studies indicated that PDX1 expression was restricted to the gastrointestinal tract. Using a cre-dependent reporter, we observed PDX1-dependent expression of tdtomato (PDX1-tom) in a subpopulation of sensory nerves. Many of these PDX1-tom afferents expressed the neurofilament 200 protein and projected to the skin. Tdtomato-labeled terminals were associated with hair follicles in the form of longitudinal and circumferential lanceolate endings suggesting a role in tactile and proprioceptive perception. To begin to examine the functional significance of PDX1 in afferents, we used Fura-2 imaging to examine calcium (Ca2+) handling under naïve and nerve injury conditions. Neuropathic injury is associated with increased intracellular Ca2+ signaling that in part results from dysregulation of the sarco/endoplasmic reticulum calcium transport ATPase (SERCA). Here we demonstrate that under naïve conditions, PDX1 regulates expression of the SERCA2B isoform in sensory neurons. In response to infraorbital nerve injury, a significant reduction of PDX1 and SERCA2B expression and dysregulation of Ca2+ handling occurs in PDX1-tom trigeminal ganglia neurons. The identification of PDX1 expression in the somatosensory system and its regulation of SERCA2B and Ca2+ handling provide a new mechanism to explain pathological changes in primary afferents that may contribute to pain associated with nerve injury.
Collapse
Affiliation(s)
- Jami L Saloman
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Ariel Y Epouhe
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Catherine F Ruff
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Neurobiology, Center for Neuroscience and Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Elkhoury K, Kodeih S, Enciso‐Martínez E, Maziz A, Bergaud C. Advancing Cardiomyocyte Maturation: Current Strategies and Promising Conductive Polymer-Based Approaches. Adv Healthc Mater 2024; 13:e2303288. [PMID: 38349615 PMCID: PMC11468390 DOI: 10.1002/adhm.202303288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/31/2024] [Indexed: 02/21/2024]
Abstract
Cardiovascular diseases are a leading cause of mortality and pose a significant burden on healthcare systems worldwide. Despite remarkable progress in medical research, the development of effective cardiovascular drugs has been hindered by high failure rates and escalating costs. One contributing factor is the limited availability of mature cardiomyocytes (CMs) for accurate disease modeling and drug screening. Human induced pluripotent stem cell-derived CMs offer a promising source of CMs; however, their immature phenotype presents challenges in translational applications. This review focuses on the road to achieving mature CMs by summarizing the major differences between immature and mature CMs, discussing the importance of adult-like CMs for drug discovery, highlighting the limitations of current strategies, and exploring potential solutions using electro-mechano active polymer-based scaffolds based on conductive polymers. However, critical considerations such as the trade-off between 3D systems and nutrient exchange, biocompatibility, degradation, cell adhesion, longevity, and integration into wider systems must be carefully evaluated. Continued advancements in these areas will contribute to a better understanding of cardiac diseases, improved drug discovery, and the development of personalized treatment strategies for patients with cardiovascular disorders.
Collapse
Affiliation(s)
- Kamil Elkhoury
- LAAS‐CNRS, Université de Toulouse, CNRSToulouseF‐31400France
| | - Sacha Kodeih
- Faculty of Medicine and Medical SciencesUniversity of BalamandTripoliP.O. Box 100Lebanon
| | | | - Ali Maziz
- LAAS‐CNRS, Université de Toulouse, CNRSToulouseF‐31400France
| | | |
Collapse
|
11
|
Pant A, Moar K, Arora TK, Maurya PK. Implication of biosignatures in the progression of endometriosis. Pathol Res Pract 2024; 254:155103. [PMID: 38237401 DOI: 10.1016/j.prp.2024.155103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 02/12/2024]
Abstract
Endometriosis is an estrogen-dependent chronic inflammatory disorder involving the placement and growth of endometrial tissue outside the uterine cavity. It is the most common multifactorial disease that affects the life quality of women in reproductive age. Due to its multicomponent nature, early diagnosis of the disease is challenging. Since many genetic, epigenetic alterations and non-genetic factors contribute to the pathology of endometriosis, devising a drug therapy that directly acts on the ectopic tissue is extremely difficult. Endometriosis is a hormone-driven disease with estrogen considered as a primary driver for the development of endometriotic lesions. This study aims to identify biosignatures involved in endometriosis with and without gonadotropin releasing hormone agonists (GnRHa). GnRHa is a short peptide analog of GnRH that causes inhibition of estrogen and androgen synthesis. Microarray based-gene expression profiling was performed on total RNA extracted from endometriotic tissue samples with and without GnRHa-treated patients already published in our previous paper. The untreated group were considered as the control. Genes were then selected for validation by quantitative real-time polymerase chain reaction (qRT-PCR). qRT-PCR analysis confirmed significant downregulation in(p < 0.05) expression of DARC (p = 0.0042), CDH1 (p = 0.0027), CDH5 (p = 0.0283), ATP2A3 (p < 0.001), RGS5 (p = 0.0032), and CD36 (p = 0.0162) in endometriosis patients treated with GnRHa analogs. Although, CTNNAL1 (p = 0.0136) also showed significant results but there was upregulation in their expression levels after GnRHa treatment. Thus, an altered expression of these genes makes them a possible candidate determinant of endometriosis treated with GnRHa.
Collapse
Affiliation(s)
- Anuja Pant
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Kareena Moar
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India
| | - Taruna K Arora
- Reproductive Biology and Maternal Child Health Division, Indian Council of Medical Research, New Delhi 110029, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh 123031, India.
| |
Collapse
|
12
|
Wang Z, Yin J, Bai M, Yang J, Jiang C, Yi X, Liu Y, Gao C. New Polyene Macrolide Compounds from Mangrove-Derived Strain Streptomyces hiroshimensis GXIMD 06359: Isolation, Antifungal Activity, and Mechanism against Talaromyces marneffei. Mar Drugs 2024; 22:38. [PMID: 38248663 PMCID: PMC10819995 DOI: 10.3390/md22010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 01/23/2024] Open
Abstract
Mangrove-derived actinomycetes represent a rich source of novel bioactive natural products in drug discovery. In this study, four new polyene macrolide antibiotics antifungalmycin B-E (1-4), along with seven known analogs (5-11), were isolated from the fermentation broth of the mangrove strain Streptomyces hiroshimensis GXIMD 06359. All compounds from this strain were purified using semi-preparative HPLC and Sephadex LH-20 gel filtration while following an antifungal activity-guided fractionation. Their structures were elucidated through spectroscopic techniques including UV, HR-ESI-MS, and NMR. These compounds exhibited broad-spectrum antifungal activity against Talaromyces marneffei with minimum inhibitory concentration (MIC) values being in the range of 2-128 μg/mL except compound 2. This is the first report of polyene derivatives produced by S. hiroshimensis as bioactive compounds against T. marneffei. In vitro studies showed that compound 1 exerted a significantly stronger antifungal activity against T. marneffei than other new compounds, and the antifungal mechanism of compound 1 may be related to the disrupted cell membrane, which causes mitochondrial dysfunction, resulting in leakage of intracellular biological components, and subsequently, cell death. Taken together, this study provides a basis for compound 1 preventing and controlling talaromycosis.
Collapse
Affiliation(s)
- Zhou Wang
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Jianglin Yin
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Nanning 530200, China
| | - Meng Bai
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Jie Yang
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Cuiping Jiang
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Xiangxi Yi
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Yonghong Liu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| | - Chenghai Gao
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China; (Z.W.); (J.Y.); (M.B.); (J.Y.); (C.J.)
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China;
| |
Collapse
|
13
|
Balakrishnan P, Arasu A, Velusamy T. Targeting altered calcium homeostasis and uncoupling protein-2 promotes sensitivity in drug-resistant breast cancer cells. J Biochem Mol Toxicol 2024; 38:e23575. [PMID: 37920924 DOI: 10.1002/jbt.23575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/15/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
Metastatic breast cancer has the highest mortality rate among women owing to its poor clinical outcomes. Metastatic tumors pose challenges for treatment through conventional surgery or radiotherapy because of their diverse organ localization and resistance to various cytotoxic agents. Chemoresistance is a significant obstacle to effective breast cancer treatment owing to cancer's heterogeneous nature. Abnormalities in intracellular calcium signaling, coupled with altered mitochondrial metabolism, play a significant role in facilitating drug resistance and contribute to therapy resistance. Uncoupling protein-2 (UCP2) is considered as a marker of chemoresistance and is believed to play a major role in promoting metabolic shifts and tumor metastasis. In this context, it is imperative to understand the roles of altered calcium signaling and metabolic switching in the development of chemotherapeutic resistance. This study investigates the roles of UCP2 and intracellular calcium signaling (Ca2+ ) in promoting chemoresistance against cisplatin. Additionally, we explored the effectiveness of combining genipin (GP, a compound that reverses UCP2-mediated chemoresistance) and thapsigargin (TG, a calcium signaling modulator) in treating highly metastatic breast cancers. Our findings indicate that both aberrant Ca2+ signaling and metabolic shifts in cancer cells contribute to developing drug-resistant phenotypes, and the combination treatment of GP and TG significantly enhances drug sensitivity in these cells. Collectively, our study underscores the potential of these drug combinations as an effective approach to overcome drug resistance in chemoresistant cancers.
Collapse
Affiliation(s)
- Pavithra Balakrishnan
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, India
| | - Ashok Arasu
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, India
| | - Thirunavukkarasu Velusamy
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, India
| |
Collapse
|
14
|
Yu A, Beck M, Merzendorfer H, Yang Q. Advances in understanding insect chitin biosynthesis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 164:104058. [PMID: 38072083 DOI: 10.1016/j.ibmb.2023.104058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
Chitin, a natural polymer of N-acetylglucosamine chains, is a principal component of the apical extracellular matrix in arthropods. Chitin microfibrils serve as structural components of natural biocomposites present in the extracellular matrix of a variety of invertebrates including sponges, molluscs, nematodes, fungi and arthropods. In this review, we summarize the frontier advances of insect chitin synthesis. More specifically, we focus on the chitin synthase (CHS), which catalyzes the key biosynthesis step. CHS is also known as an attractive insecticidal target in that this enzyme is absent in mammals, birds or plants. As no insect chitin synthase structure have been reported so far, we review recent studies on glycosyltransferase domain structures derived from fungi and oomycetes, which are conserved in CHS from all species containing chitin. Auxiliary proteins, which coordinate with CHS in chitin biosynthesis and assembly, are also discussed.
Collapse
Affiliation(s)
- Ailing Yu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China; State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Marius Beck
- Department of Chemistry-Biology, University of Siegen, Siegen, Germany
| | - Hans Merzendorfer
- Department of Chemistry-Biology, University of Siegen, Siegen, Germany.
| | - Qing Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China; State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
15
|
Šeflová J, Cruz-Cortés C, Guerrero-Serna G, Robia SL, Espinoza-Fonseca LM. Mechanisms for cardiac calcium pump activation by its substrate and a synthetic allosteric modulator using fluorescence lifetime imaging. PNAS NEXUS 2024; 3:pgad453. [PMID: 38222469 PMCID: PMC10785037 DOI: 10.1093/pnasnexus/pgad453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024]
Abstract
The discovery of allosteric modulators is an emerging paradigm in drug discovery, and signal transduction is a subtle and dynamic process that is challenging to characterize. We developed a time-correlated single photon-counting imaging approach to investigate the structural mechanisms for small-molecule activation of the cardiac sarcoplasmic reticulum Ca2+-ATPase, a pharmacologically important pump that transports Ca2+ at the expense of adenosine triphosphate (ATP) hydrolysis. We first tested whether the dissociation of sarcoplasmic reticulum Ca2+-ATPase from its regulatory protein phospholamban is required for small-molecule activation. We found that CDN1163, a validated sarcoplasmic reticulum Ca2+-ATPase activator, does not have significant effects on the stability of the sarcoplasmic reticulum Ca2+-ATPase-phospholamban complex. Time-correlated single photon-counting imaging experiments using the nonhydrolyzable ATP analog β,γ-Methyleneadenosine 5'-triphosphate (AMP-PCP) showed ATP is an allosteric modulator of sarcoplasmic reticulum Ca2+-ATPase, increasing the fraction of catalytically competent structures at physiologically relevant Ca2+ concentrations. Unlike ATP, CDN1163 alone has no significant effects on the Ca2+-dependent shifts in the structural populations of sarcoplasmic reticulum Ca2+-ATPase, and it does not increase the pump's affinity for Ca2+ ions. However, we found that CDN1163 enhances the ATP-mediated modulatory effects to increase the population of catalytically competent sarcoplasmic reticulum Ca2+-ATPase structures. Importantly, this structural shift occurs within the physiological window of Ca2+ concentrations at which sarcoplasmic reticulum Ca2+-ATPase operates. We demonstrated that ATP is both a substrate and modulator of sarcoplasmic reticulum Ca2+-ATPase and showed that CDN1163 and ATP act synergistically to populate sarcoplasmic reticulum Ca2+-ATPase structures that are primed for phosphorylation. This study provides novel insights into the structural mechanisms for sarcoplasmic reticulum Ca2+-ATPase activation by its substrate and a synthetic allosteric modulator.
Collapse
Affiliation(s)
- Jaroslava Šeflová
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA
| | - Carlos Cruz-Cortés
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guadalupe Guerrero-Serna
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seth L Robia
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL 60153, USA
| | - L Michel Espinoza-Fonseca
- Center for Arrhythmia Research, Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Qian MF, Bevacqua RJ, Coykendall VM, Liu X, Zhao W, Chang CA, Gu X, Dai XQ, MacDonald PE, Kim SK. HNF1α maintains pancreatic α and β cell functions in primary human islets. JCI Insight 2023; 8:e170884. [PMID: 37943614 PMCID: PMC10807710 DOI: 10.1172/jci.insight.170884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
HNF1A haploinsufficiency underlies the most common form of human monogenic diabetes (HNF1A-maturity onset diabetes of the young [HNF1A-MODY]), and hypomorphic HNF1A variants confer type 2 diabetes risk. But a lack of experimental systems for interrogating mature human islets has limited our understanding of how the transcription factor HNF1α regulates adult islet function. Here, we combined conditional genetic targeting in human islet cells, RNA-Seq, chromatin mapping with cleavage under targets and release using nuclease (CUT&RUN), and transplantation-based assays to determine HNF1α-regulated mechanisms in adult human pancreatic α and β cells. Short hairpin RNA-mediated (shRNA-mediated) suppression of HNF1A in primary human pseudoislets led to blunted insulin output and dysregulated glucagon secretion after transplantation in mice, recapitulating phenotypes observed in patients with diabetes. These deficits corresponded with altered expression of genes encoding factors critical for hormone secretion, including calcium channel subunits, ATPase transporters, and extracellular matrix constituents. Additionally, HNF1A loss led to upregulation of transcriptional repressors, providing evidence for a mechanism of transcriptional derepression through HNF1α. CUT&RUN mapping of HNF1α DNA binding sites in primary human islets imputed a subset of HNF1α-regulated genes as direct targets. These data elucidate mechanistic links between HNF1A loss and diabetic phenotypes in mature human α and β cells.
Collapse
Affiliation(s)
- Mollie F. Qian
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Romina J. Bevacqua
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vy M.N. Coykendall
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Xiong Liu
- Department of Pharmacology and
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Weichen Zhao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Charles A. Chang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Xueying Gu
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
- Stanford Diabetes Research Center
| | - Xiao-Qing Dai
- Department of Pharmacology and
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Patrick E. MacDonald
- Department of Pharmacology and
- Alberta Diabetes Institute, University of Alberta, Edmonton, Canada
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
- Stanford Diabetes Research Center
- Departments of Medicine and Pediatrics (Endocrinology), and
- Northern California JDRF Center of Excellence, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
17
|
Lu Y, Ran Y, Li H, Wen J, Cui X, Zhang X, Guan X, Cheng M. Micropeptides: origins, identification, and potential role in metabolism-related diseases. J Zhejiang Univ Sci B 2023; 24:1106-1122. [PMID: 38057268 PMCID: PMC10710913 DOI: 10.1631/jzus.b2300128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/06/2023] [Indexed: 12/08/2023]
Abstract
With the development of modern sequencing techniques and bioinformatics, genomes that were once thought to be noncoding have been found to encode abundant functional micropeptides (miPs), a kind of small polypeptides. Although miPs are difficult to analyze and identify, a number of studies have begun to focus on them. More and more miPs have been revealed as essential for energy metabolism homeostasis, immune regulation, and tumor growth and development. Many reports have shown that miPs are especially essential for regulating glucose and lipid metabolism and regulating mitochondrial function. MiPs are also involved in the progression of related diseases. This paper reviews the sources and identification of miPs, as well as the functional significance of miPs for metabolism-related diseases, with the aim of revealing their potential clinical applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Min Cheng
- School of Basic Medicine Sciences, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
18
|
Abdel-Razek O, Marzouk A, MacKinnon M, Guy ET, Pohar SA, Zhushma E, Liu J, Sia I, Gokey JJ, Tay HG, Amack JD. Calcium signaling mediates proliferation of the precursor cells that give rise to the ciliated left-right organizer in the zebrafish embryo. Front Mol Biosci 2023; 10:1292076. [PMID: 38152112 PMCID: PMC10751931 DOI: 10.3389/fmolb.2023.1292076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 12/29/2023] Open
Abstract
Several of our internal organs, including heart, lungs, stomach, and spleen, develop asymmetrically along the left-right (LR) body axis. Errors in establishing LR asymmetry, or laterality, of internal organs during early embryonic development can result in birth defects. In several vertebrates-including humans, mice, frogs, and fish-cilia play a central role in establishing organ laterality. Motile cilia in a transient embryonic structure called the "left-right organizer" (LRO) generate a directional fluid flow that has been proposed to be detected by mechanosensory cilia to trigger asymmetric signaling pathways that orient the LR axis. However, the mechanisms that control the form and function of the ciliated LRO remain poorly understood. In the zebrafish embryo, precursor cells called dorsal forerunner cells (DFCs) develop into a transient ciliated structure called Kupffer's vesicle (KV) that functions as the LRO. DFCs can be visualized and tracked in the embryo, thereby providing an opportunity to investigate mechanisms that control LRO development. Previous work revealed that proliferation of DFCs via mitosis is a critical step for developing a functional KV. Here, we conducted a targeted pharmacological screen to identify mechanisms that control DFC proliferation. Small molecule inhibitors of the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) were found to reduce DFC mitosis. The SERCA pump is involved in regulating intracellular calcium ion (Ca2+) concentration. To visualize Ca2+ in living embryos, we generated transgenic zebrafish using the fluorescent Ca2+ biosensor GCaMP6f. Live imaging identified dynamic cytoplasmic Ca2+ transients ("flux") that occur unambiguously in DFCs. In addition, we report Ca2+ flux events that occur in the nucleus of DFCs. Nuclear Ca2+ flux occurred in DFCs that were about to undergo mitosis. We find that SERCA inhibitor treatments during DFC proliferation stages alters Ca2+ dynamics, reduces the number of ciliated cells in KV, and alters embryo laterality. Mechanistically, SERCA inhibitor treatments eliminated both cytoplasmic and nuclear Ca2+ flux events, and reduced progression of DFCs through the S/G2 phases of the cell cycle. These results identify SERCA-mediated Ca2+ signaling as a mitotic regulator of the precursor cells that give rise to the ciliated LRO.
Collapse
Affiliation(s)
- Osama Abdel-Razek
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Amanda Marzouk
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Madison MacKinnon
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Edward T. Guy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Sonny A. Pohar
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Emily Zhushma
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Junjie Liu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Isabel Sia
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jason J. Gokey
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Hwee Goon Tay
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
| | - Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, NY, United States
| |
Collapse
|
19
|
Chen B, Tan L, Wang Y, Yang L, Liu J, Chen D, Huang S, Mao F, Lian J. LOC102549726/miR-760-3p network is involved in the progression of ISO-induced pathological cardiomyocyte hypertrophy via endoplasmic reticulum stress. J Mol Histol 2023; 54:675-687. [PMID: 37899367 PMCID: PMC10635935 DOI: 10.1007/s10735-023-10166-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 09/30/2023] [Indexed: 10/31/2023]
Abstract
Pathological cardiac hypertrophy (CH) is featured by myocyte enlargement and cardiac malfunction. Multiple signaling pathways have been implicated in diverse pathological and physiological processes in CH. However, the function of LOC102549726/miR-760-3p network in CH remains unclear. Here, we characterize the functional role of LOC102549726/miR-760-3p network in CH and delineate the underlying mechanism. The expression of LncRNA LOC102549726 and hypertrophic markers was significantly increased compared to the control, while the level of miR-760-3p was decreased. Next, we examined ER stress response in a hypertrophic cardiomyocyte model. The expression of ER stress markers was greatly enhanced after incubation with ISO. The hypertrophic reaction, ER stress response, and increased potassium and calcium ion channels were alleviated by genetic downregulation of LOC102549726. It has been demonstrated that LOC102549726 functions as a competitive endogenous RNA (ceRNA) of miR-760-3p. Overexpression of miR-760-3p decreased cell surface area and substantially mitigated ER stress response; protein levels of potassium and calcium channels were also significantly up-regulated compared to the NC control. In contrast, miR-760-3p inhibition increased cell size, aggravated CH and ER stress responses, and reduced ion channels. Collectively, in this study we demonstrated that the LOC102549726/miR-760-3p network was a crucial regulator of CH development. Ion channels mediate the ER stress response and may be a downstream sensor of the LOC102549726/miR-760-3p network. Therefore, these findings advance our understanding of pathological CH and provide new insights into therapeutic targets for cardiac remodeling.
Collapse
Affiliation(s)
- Bangsheng Chen
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Lian Tan
- Intensive Care Unit, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Ying Wang
- Cadiovascular Department, Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, 315100, China
| | - Lei Yang
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Jiequan Liu
- Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Danqi Chen
- Intensive Care Unit, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Shuaishuai Huang
- Laboratory of Renal Carcinoma, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, 315192, China
| | - Feiyan Mao
- Department of General Surgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, 315100, China
| | - Jiangfang Lian
- Cadiovascular Department, Ningbo Medical Center LiHuiLi Hospital, Ningbo, Zhejiang, 315100, China.
| |
Collapse
|
20
|
Tarfeen N, Nisa KU, Ahmad MB, Waza AA, Ganai BA. Metabolic and Genetic Association of Vitamin D with Calcium Signaling and Insulin Resistance. Indian J Clin Biochem 2023; 38:407-417. [PMID: 37746541 PMCID: PMC10516840 DOI: 10.1007/s12291-022-01105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Various evidences have unveiled the significance of Vitamin D in diverse processes which include its action in prevention of immune dysfunction, cancer and cardiometabolic disorders. Studies have confirmed the function of VD in controlling the expression of approximately nine hundred genes including gene expression of insulin. VD insufficiency may be linked with the pathogenesis of diseases that are associated with insulin resistance (IR) including diabetes as well as obesity. Thus, VD lowers IR-related disorders such as inflammation and oxidative stress. This review provides an insight regarding the molecular mechanism manifesting, how insufficiency of VD may be connected with the IR and diabetes. It also discusses the effect of VD in maintaining the Ca2+ levels in beta cells of the pancreas and in the tissues that are responsive to insulin.
Collapse
Affiliation(s)
- Najeebul Tarfeen
- Centre of Research for Development, University of Kashmir, Srinagar, India
| | - Khair Ul Nisa
- Department of Environmental Science, University of Kashmir, Srinagar, India
| | - Mir Bilal Ahmad
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Ajaz Ahmad Waza
- Multidisciplinary Research Unit (MRU), Government Medical Collage (GMC) Srinagar, Srinagar, J & K 190010 India
| | - Bashir Ahmad Ganai
- Centre of Research for Development, University of Kashmir, Srinagar, India
| |
Collapse
|
21
|
Sarkar P, Jassar O, Ghanim M. The plant pathogenic bacterium Candidatus Liberibacter solanacearum induces calcium-regulated autophagy in midgut cells of its insect vector Bactericera trigonica. Microbiol Spectr 2023; 11:e0130123. [PMID: 37768086 PMCID: PMC10581152 DOI: 10.1128/spectrum.01301-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 08/11/2023] [Indexed: 09/29/2023] Open
Abstract
Autophagy plays an important role against pathogen infection in many organisms; however, little has been done with regard to vector-borne plant and animal pathogens, that sometimes replicate and cause deleterious effects in their vectors. Candidatus Liberibacter solanacearum (CLso) is a fastidious gram-negative phloem-restricted plant pathogen and vectored by the carrot psyllid, Bactericera trigonica. The plant disease caused by this bacterium is called carrot yellows and has recently gained much importance due to worldwide excessive economical losses. Here, we demonstrate that calcium ATPase, cytosolic calcium, and most importantly Beclin-1 have a role in regulating autophagy and its association with Liberibacter inside the psyllid. The presence of CLso generates reactive oxygen species and induces the expression of detoxification enzymes in the psyllid midguts, a main site for bacteria transmission. CLso also induces the expression of both sarco/endoplasmic reticulum Ca2+pump (SERCA) and 1,4,5-trisphosphate receptors (ITPR) in midguts, resulting in high levels of calcium in the cellular cytosol. Silencing these genes individually disrupted the calcium levels in the cytosol and resulted in direct effects on autophagy and subsequently on Liberibacter persistence and transmission. Inhibiting Beclin1-phosphorylation through different calcium-induced kinases altered the expression of autophagy and CLso titers and persistence. Based on our results obtained from the midgut, we suggest the existence of a direct correlation between cytosolic calcium levels, autophagy, and CLso persistence and transmission by the carrot psyllid. IMPORTANCE Plant diseases caused by vector-borne Liberibacter species are responsible for the most important economic losses in many agricultural sectors. Preventing these diseases relies mostly on chemical sprays against the insect vectors. Knowledge-based interference with the bacteria-vector interaction remains a promising approach as a sustainable solution. For unravelling how Liberibacter exploits molecular pathways in its insect vector for transmission, here, we show that the bacterium manipulates calcium levels on both sides of the endoplasmic reticulum membrane, resulting in manipulating autophagy. Silencing genes associated with these pathways disrupted the calcium levels in the cytosol and resulted in direct effects on autophagy and Liberibacter transmission. These results demonstrate major pathways that could be exploited for manipulating and controlling the disease transmission.
Collapse
Affiliation(s)
- Poulami Sarkar
- Department of Entomology, Volcani Institute, Rishon LeZion, Israel
| | - Ola Jassar
- Department of Entomology, Volcani Institute, Rishon LeZion, Israel
- Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Murad Ghanim
- Department of Entomology, Volcani Institute, Rishon LeZion, Israel
| |
Collapse
|
22
|
Kim JM, Dziobaka S, Yoon YE, Lee HL, Jeong JH, Lee IR, Weidinger D, Yang C, Kim D, Gulperi Y, Lee CK, Sohn J, Song G, Hatt H, Lee SJ. OR2H2 Activates CAMKKβ-AMPK-Autophagy Signaling Axis and Suppresses Senescence in VK2/E6E7 Cells. Pharmaceuticals (Basel) 2023; 16:1221. [PMID: 37765029 PMCID: PMC10535153 DOI: 10.3390/ph16091221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/11/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Olfactory receptors are expressed in multiple extra-nasal tissues and these ectopic olfactory receptors mediate tissue-specific functions and regulate cellular physiology. Ectopic olfactory receptors may play key roles in tissues constantly exposed to odorants, thus the functionality of these receptors in genital tissues is of particular interest. The functionality of ectopic olfactory receptors expressed in VK2/E6E7 human vaginal epithelial cells was investigated. OR2H2 was the most highly expressed olfactory receptor expressed in VK2/E6E7 cells, and activation of OR2H2 by aldehyde 13-13, a ligand of OR2H2, increased the intracellular calcium and cAMP concentrations. Immunoblotting demonstrated that activation of OR2H2 by aldehyde 13-13 stimulated the CAMKKβ-AMPK-mTORC1-autophagy signaling axis, and that these effects were negated by OR2H2 knockdown. AMPK is known to regulate senescence; consequently, we investigated further the effect of aldehyde 13-13 on senescence. In H2O2-induced senescent cells, activation of OR2H2 by aldehyde 13-13 restored proliferation, and reduced the expression of senescence markers, P16 and P19. Additionally, aldehyde 13-13 induced apoptosis of H2O2-induced senescent cells, compared with non-senescent normal cells. In vivo, aldehyde 13-13 increased the lifespan of Caenorhabditis elegans and budding yeast. These findings demonstrate that OR2H2 is a functional receptor in VK2/E6E7 cells, and that activation of OR2H2 activates the AMPK-autophagy axis, and suppresses cellular aging and senescence, which may increase cellular health.
Collapse
Affiliation(s)
- Ji Min Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
| | - Sina Dziobaka
- Department of Cell Physiology, Ruhr-University Bochum, 44791 Bochum, Germany; (S.D.); (D.W.)
| | - Ye Eun Yoon
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
| | - Ha Lim Lee
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
| | - Ji Hyun Jeong
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
| | - In-Ryeong Lee
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
| | - Daniel Weidinger
- Department of Cell Physiology, Ruhr-University Bochum, 44791 Bochum, Germany; (S.D.); (D.W.)
| | - Changwon Yang
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
| | - Deokho Kim
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
| | - Yalcin Gulperi
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
| | - Cheol-Koo Lee
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
| | - Jeongwon Sohn
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 02842, Republic of Korea;
- Korea Institute of Molecular Medicine and Nutrition, Seoul 02842, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, School of Life Science and Biotechnology for BK21 PLUS, Korea University, Seoul 02855, Republic of Korea; (J.M.K.); (Y.E.Y.); (H.L.L.); (J.H.J.); (I.-R.L.); (C.Y.); (D.K.); (Y.G.); (C.-K.L.)
- Division of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
| | - Hanns Hatt
- Department of Cell Physiology, Ruhr-University Bochum, 44791 Bochum, Germany; (S.D.); (D.W.)
| | - Sung-Joon Lee
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul 02855, Republic of Korea
- Interdisciplinary Program in Precision Public Health, Korea University, Seoul 02846, Republic of Korea
- BK21 Four Institute of Precision Public Health, Korea University, Seoul 02846, Republic of Korea
| |
Collapse
|
23
|
Yang S, Xie BL, Dong XP, Wang LX, Zhu GH, Wang T, Wu WJ, Lai RS, Tao R, Guan MX, Chen FY, Tan DH, Deng Z, Xie HP, Zeng Y, Xiao ZA, Xie DH. cdh23 affects congenital hearing loss through regulating purine metabolism. Front Mol Neurosci 2023; 16:1079529. [PMID: 37575969 PMCID: PMC10416109 DOI: 10.3389/fnmol.2023.1079529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/13/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction The pathogenic gene CDH23 plays a pivotal role in tip links, which is indispensable for mechanoelectrical transduction in the hair cells. However, the underlying molecular mechanism and signal regulatory networks that influence deafness is still largely unknown. Methods In this study, a congenital deafness family, whole exome sequencing revealed a new mutation in the pathogenic gene CDH23, subsequently; the mutation has been validated using Sanger sequencing method. Then CRISPR/Cas9 technology was employed to knockout zebrafish cdh23 gene. Startle response experiment was used to compare with wide-type, the response to sound stimulation between wide-type and cdh23-/-. To further illustrate the molecular mechanisms underlying congenital deafness, comparative transcriptomic profiling and multiple bioinformatics analyses were performed. Results The YO-PRO-1 assay result showed that in cdh23 deficient embryos, the YO-PRO-1 signal in inner ear and lateral line neuromast hair cells were completely lost. Startle response experiment showed that compared with wide-type, the response to sound stimulation decreased significantly in cdh23 mutant larvae. Comparative transcriptomic showed that the candidate genes such as atp1b2b and myof could affect hearing by regulating ATP production and purine metabolism in a synergetic way with cdh23. RT-qPCR results further confirmed the transcriptomics results. Further compensatory experiment showed that ATP treated cdh23-/- embryos can partially recover the mutant phenotype. Conclusion In conclusion, our study may shed light on deciphering the principal mechanism and provide a potential therapeutic method for congenital hearing loss under the condition of CDH23 mutation.
Collapse
Affiliation(s)
- Shu Yang
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Bing-Lin Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Xiao-ping Dong
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Ling-xiang Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Gang-hua Zhu
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Tian Wang
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei-jing Wu
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ruo-sha Lai
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Rong Tao
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min-xin Guan
- Institute of Genetics, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Human Genetics, Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Fang-yi Chen
- Shenzhen Key Laboratory of Smart Healthcare Engineering, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Dong-hui Tan
- Department of Otolaryngology—Head and Neck Surgery, The Affiliated Hospital of Xiang Nan College, Chenzhou, China
| | - Zhong Deng
- Department of Otolaryngology—Head and Neck Surgery, The Affiliated Hospital of Xiang Nan College, Chenzhou, China
| | - Hua-ping Xie
- Laboratory of Animal Nutrition and Human Health, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Yong Zeng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Changsha, Hunan, China
| | - Zi-an Xiao
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ding-hua Xie
- Department of Otorhinolaryngology—Head & Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Adamo FM, Silva Barcelos EC, De Falco F, Dorillo E, Rompietti C, Sorcini D, Stella A, Del Papa B, Baldoni S, Esposito A, Geraci C, Arcaleni R, Pennetta C, Ragonese F, Moretti L, Mameli M, Di Ianni M, Rosati E, Fioretti B, Sportoletti P. Therapeutic Targeting Potential of Novel Silver Nanoparticles Coated with Anti-CD20 Antibody against Chronic Lymphocytic Leukemia. Cancers (Basel) 2023; 15:3618. [PMID: 37509279 PMCID: PMC10377400 DOI: 10.3390/cancers15143618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) is an incurable disorder associated with alterations in several pathways essential for survival and proliferation. Despite the advances made in CLL therapy with the new target agents, in some cases, relapses and resistance could occur, making the discovery of new alternatives to manage CLL refractoriness necessary. To provide new therapeutic strategies for CLL, we investigated the anti-leukemic activity of silver nanoparticles (AgNPs), whose impact on CLL cells has been poorly explored. METHODS We studied the action mechanisms of AgNPs in vitro through flow cytometry and molecular analyses. To improve the bioavailability of AgNPs, we generated AgNPs coated with the anti-CD20 antibody Rituximab (AgNPs@Rituximab) and carried out imaging-based approaches and in vivo experiments to evaluate specificity, drug uptake, and efficacy. RESULTS AgNPs reduced the viability of primary CLL cells and the HG-3 cell line by inducing an intrinsic apoptotic pathway characterized by Bax/Bcl-2 imbalance, caspase activation, and PARP degradation. Early apoptotic events triggered by AgNPs included enhanced Ca2+ influx and ROS overproduction. AgNPs synergistically potentiated the cytotoxicity of Venetoclax, Ibrutinib, and Bepridil. In vitro, the AgNPs@Rituximab conjugates were rapidly internalized within CLL cells and strongly prolonged the survival of CLL xenograft models compared to each unconjugated single agent. CONCLUSIONS AgNPs showed strong anti-leukemic activity in CLL, with the potential for clinical translation in combination with agents used in CLL. The increased specificity of AgNPs@Rituximab toward CLL cells could be relevant for overcoming in vivo AgNPs' non-specific distribution and increasing their efficacy.
Collapse
Affiliation(s)
- Francesco Maria Adamo
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Estevao Carlos Silva Barcelos
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
- Postgraduate Program in Biotechnology, Federal University of Espírito Santo, Vitória 29043-900, Brazil
| | - Filomena De Falco
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Erica Dorillo
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Chiara Rompietti
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Daniele Sorcini
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Arianna Stella
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Beatrice Del Papa
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Stefano Baldoni
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Angela Esposito
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Clelia Geraci
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Roberta Arcaleni
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Chiara Pennetta
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy
| | - Lorenzo Moretti
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Mariagrazia Mameli
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| | - Mauro Di Ianni
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Emanuela Rosati
- Department of Medicine and Surgery, Biosciences and Medical Embryology Section, University of Perugia, 06129 Perugia, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, 06123 Perugia, Italy
| | - Paolo Sportoletti
- Department of Medicine and Surgery, Institute of Hematology and Center for Hemato-Oncology Research (CREO), University of Perugia, Santa Maria della Misericordia Hospital, 06129 Perugia, Italy
| |
Collapse
|
25
|
Plasterer C, Semenikhina M, Tsaih SW, Flister MJ, Palygin O. NNAT is a novel mediator of oxidative stress that suppresses ER + breast cancer. Mol Med 2023; 29:87. [PMID: 37400769 PMCID: PMC10318825 DOI: 10.1186/s10020-023-00673-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/30/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Neuronatin (NNAT) was recently identified as a novel mediator of estrogen receptor-positive (ER+) breast cancer cell proliferation and migration, which correlated with decreased tumorigenic potential and prolonged patient survival. However, despite these observations, the molecular and pathophysiological role(s) of NNAT in ER + breast cancer remains unclear. Based on high protein homology with phospholamban, we hypothesized that NNAT mediates the homeostasis of intracellular calcium [Ca2+]i levels and endoplasmic reticulum (EndoR) function, which is frequently disrupted in ER + breast cancer and other malignancies. METHODS To evaluate the role of NNAT on [Ca2+]i homeostasis, we used a combination of bioinformatics, gene expression and promoter activity assays, CRISPR gene manipulation, pharmacological tools and confocal imaging to characterize the association between ROS, NNAT and calcium signaling. RESULTS Our data indicate that NNAT localizes predominantly to EndoR and lysosome, and genetic manipulation of NNAT levels demonstrated that NNAT modulates [Ca2+]i influx and maintains Ca2+ homeostasis. Pharmacological inhibition of calcium channels revealed that NNAT regulates [Ca2+]i levels in breast cancer cells through the interaction with ORAI but not the TRPC signaling cascade. Furthermore, NNAT is transcriptionally regulated by NRF1, PPARα, and PPARγ and is strongly upregulated by oxidative stress via the ROS and PPAR signaling cascades. CONCLUSION Collectively, these data suggest that NNAT expression is mediated by oxidative stress and acts as a regulator of Ca2+ homeostasis to impact ER + breast cancer proliferation, thus providing a molecular link between the longstanding observation that is accumulating ROS and altered Ca2+ signaling are key oncogenic drivers of cancer.
Collapse
Affiliation(s)
- Cody Plasterer
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Marharyta Semenikhina
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shirng-Wern Tsaih
- Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael J Flister
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, USA.
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA.
| |
Collapse
|
26
|
Dowling P, Swandulla D, Ohlendieck K. Biochemical and proteomic insights into sarcoplasmic reticulum Ca 2+-ATPase complexes in skeletal muscles. Expert Rev Proteomics 2023; 20:125-142. [PMID: 37668143 DOI: 10.1080/14789450.2023.2255743] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/07/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Skeletal muscles contain large numbers of high-molecular-mass protein complexes in elaborate membrane systems. Integral membrane proteins are involved in diverse cellular functions including the regulation of ion handling, membrane homeostasis, energy metabolism and force transmission. AREAS COVERED The proteomic profiling of membrane proteins and large protein assemblies in skeletal muscles are outlined in this article. This includes a critical overview of the main biochemical separation techniques and the mass spectrometric approaches taken to study membrane proteins. As an illustrative example of an analytically challenging large protein complex, the proteomic detection and characterization of the Ca2+-ATPase of the sarcoplasmic reticulum is discussed. The biological role of this large protein complex during normal muscle functioning, in the context of fiber type diversity and in relation to mechanisms of physiological adaptations and pathophysiological abnormalities is evaluated from a proteomics perspective. EXPERT OPINION Mass spectrometry-based muscle proteomics has decisively advanced the field of basic and applied myology. Although it is technically challenging to study membrane proteins, innovations in protein separation methodology in combination with sensitive mass spectrometry and improved systems bioinformatics has allowed the detailed proteomic detection and characterization of skeletal muscle membrane protein complexes, such as Ca2+-pump proteins of the sarcoplasmic reticulum.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth Kildare, Ireland
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth Kildare, Ireland
| |
Collapse
|
27
|
Jurcau A, Jurcau CM. Mitochondria in Huntington's disease: implications in pathogenesis and mitochondrial-targeted therapeutic strategies. Neural Regen Res 2023; 18:1472-1477. [PMID: 36571344 PMCID: PMC10075114 DOI: 10.4103/1673-5374.360289] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Huntington's disease is a genetic disease caused by expanded CAG repeats on exon 1 of the huntingtin gene located on chromosome 4. Compelling evidence implicates impaired mitochondrial energetics, altered mitochondrial biogenesis and quality control, disturbed mitochondrial trafficking, oxidative stress and mitochondrial calcium dyshomeostasis in the pathogenesis of the disorder. Unfortunately, conventional mitochondrial-targeted molecules, such as cysteamine, creatine, coenzyme Q10, or triheptanoin, yielded negative or inconclusive results. However, future therapeutic strategies, aiming to restore mitochondrial biogenesis, improving the fission/fusion balance, and improving mitochondrial trafficking, could prove useful tools in improving the phenotype of Huntington's disease and, used in combination with genome-editing methods, could lead to a cure for the disease.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea; Neurology 3 Ward, Clinical Emergency Hospital, Oradea, Romania
| | | |
Collapse
|
28
|
Svarcbahs R, Blossom SM, Baffoe-Bonnie HS, Trychta KA, Greer LK, Pickel J, Henderson MJ, Harvey BK. A transgenic mouse line for assaying tissue-specific changes in endoplasmic reticulum proteostasis. Transgenic Res 2023; 32:209-221. [PMID: 37133648 PMCID: PMC10195735 DOI: 10.1007/s11248-023-00349-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/17/2023] [Indexed: 05/04/2023]
Abstract
Maintenance of calcium homeostasis is important for proper endoplasmic reticulum (ER) function. When cellular stress conditions deplete the high concentration of calcium in the ER, ER-resident proteins are secreted into the extracellular space in a process called exodosis. Monitoring exodosis provides insight into changes in ER homeostasis and proteostasis resulting from cellular stress associated with ER calcium dysregulation. To monitor cell-type specific exodosis in the intact animal, we created a transgenic mouse line with a Gaussia luciferase (GLuc)-based, secreted ER calcium-modulated protein, SERCaMP, preceded by a LoxP-STOP-LoxP (LSL) sequence. The Cre-dependent LSL-SERCaMP mice were crossed with albumin (Alb)-Cre and dopamine transporter (DAT)-Cre mouse lines. GLuc-SERCaMP expression was characterized in mouse organs and extracellular fluids, and the secretion of GLuc-SERCaMP in response to cellular stress was monitored following pharmacological depletion of ER calcium. In LSL-SERCaMP × Alb-Cre mice, robust GLuc activity was observed only in the liver and blood, whereas in LSL-SERCaMP × DAT-Cre mice, GLuc activity was seen in midbrain dopaminergic neurons and tissue samples innervated by dopaminergic projections. After calcium depletion, we saw increased GLuc signal in the plasma and cerebrospinal fluid collected from the Alb-Cre and DAT-Cre crosses, respectively. This mouse model can be used to investigate the secretion of ER-resident proteins from specific cell and tissue types during disease pathogenesis and may aid in the identification of therapeutics and biomarkers of disease.
Collapse
Affiliation(s)
- Reinis Svarcbahs
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sarah M Blossom
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Helena S Baffoe-Bonnie
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kathleen A Trychta
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lacey K Greer
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - James Pickel
- Transgenic Technology Core, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Mark J Henderson
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Brandon K Harvey
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
29
|
Ghosh N, Chacko L, Bhattacharya H, Vallamkondu J, Nag S, Dey A, Karmakar T, Reddy PH, Kandimalla R, Dewanjee S. Exploring the Complex Relationship between Diabetes and Cardiovascular Complications: Understanding Diabetic Cardiomyopathy and Promising Therapies. Biomedicines 2023; 11:biomedicines11041126. [PMID: 37189744 DOI: 10.3390/biomedicines11041126] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
Diabetes mellitus (DM) and cardiovascular complications are two unmet medical emergencies that can occur together. The rising incidence of heart failure in diabetic populations, in addition to apparent coronary heart disease, ischemia, and hypertension-related complications, has created a more challenging situation. Diabetes, as a predominant cardio-renal metabolic syndrome, is related to severe vascular risk factors, and it underlies various complex pathophysiological pathways at the metabolic and molecular level that progress and converge toward the development of diabetic cardiomyopathy (DCM). DCM involves several downstream cascades that cause structural and functional alterations of the diabetic heart, such as diastolic dysfunction progressing into systolic dysfunction, cardiomyocyte hypertrophy, myocardial fibrosis, and subsequent heart failure over time. The effects of glucagon-like peptide-1 (GLP-1) analogues and sodium-glucose cotransporter-2 (SGLT-2) inhibitors on cardiovascular (CV) outcomes in diabetes have shown promising results, including improved contractile bioenergetics and significant cardiovascular benefits. The purpose of this article is to highlight the various pathophysiological, metabolic, and molecular pathways that contribute to the development of DCM and its significant effects on cardiac morphology and functioning. Additionally, this article will discuss the potential therapies that may be available in the future.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Molecular Pharmacology Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, Rockville, MD 20850-3173, USA
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | | | - Sagnik Nag
- Department of Biotechnology, Vellore Institute of Technology (VIT), School of Biosciences & Technology, Tiruvalam Road, Vellore 632014, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, India
| | - Tanushree Karmakar
- Dr. B C Roy College of Pharmacy and Allied Health Sciences, Durgapur 713206, India
| | | | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
30
|
Marchioretti C, Zanetti G, Pirazzini M, Gherardi G, Nogara L, Andreotti R, Martini P, Marcucci L, Canato M, Nath SR, Zuccaro E, Chivet M, Mammucari C, Pacifici M, Raffaello A, Rizzuto R, Mattarei A, Desbats MA, Salviati L, Megighian A, Sorarù G, Pegoraro E, Belluzzi E, Pozzuoli A, Biz C, Ruggieri P, Romualdi C, Lieberman AP, Babu GJ, Sandri M, Blaauw B, Basso M, Pennuto M. Defective excitation-contraction coupling and mitochondrial respiration precede mitochondrial Ca 2+ accumulation in spinobulbar muscular atrophy skeletal muscle. Nat Commun 2023; 14:602. [PMID: 36746942 PMCID: PMC9902403 DOI: 10.1038/s41467-023-36185-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/19/2023] [Indexed: 02/08/2023] Open
Abstract
Polyglutamine expansion in the androgen receptor (AR) causes spinobulbar muscular atrophy (SBMA). Skeletal muscle is a primary site of toxicity; however, the current understanding of the early pathological processes that occur and how they unfold during disease progression remains limited. Using transgenic and knock-in mice and patient-derived muscle biopsies, we show that SBMA mice in the presymptomatic stage develop a respiratory defect matching defective expression of genes involved in excitation-contraction coupling (ECC), altered contraction dynamics, and increased fatigue. These processes are followed by stimulus-dependent accumulation of calcium into mitochondria and structural disorganization of the muscle triads. Deregulation of expression of ECC genes is concomitant with sexual maturity and androgen raise in the serum. Consistent with the androgen-dependent nature of these alterations, surgical castration and AR silencing alleviate the early and late pathological processes. These observations show that ECC deregulation and defective mitochondrial respiration are early but reversible events followed by altered muscle force, calcium dyshomeostasis, and dismantling of triad structure.
Collapse
Affiliation(s)
- Caterina Marchioretti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Giulia Zanetti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Marco Pirazzini
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Gaia Gherardi
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Roberta Andreotti
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, 25121, Brescia, Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Marta Canato
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Samir R Nath
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Emanuela Zuccaro
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Mathilde Chivet
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Cristina Mammucari
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Marco Pacifici
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Anna Raffaello
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
| | - Andrea Mattarei
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Maria A Desbats
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Leonardo Salviati
- CIR-Myo, Centro Interdipartimentale di Ricerca di Miologia, University of Padova, 35131, Padova, Italy
- Clinical Genetics Unit, Department of Women and Children's Health, University of Padova, and Fondazione Istituto di Ricerca Pediatrica Città della Speranza, Padova, Italy
| | - Aram Megighian
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
| | - Gianni Sorarù
- Padova Neuroscience Center (PNC), Padova, 35100, Italy
- Department of Neuroscience (DNS), University of Padova, 35128, Padova, Italy
| | - Elena Pegoraro
- Department of Neuroscience (DNS), University of Padova, 35128, Padova, Italy
| | - Elisa Belluzzi
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128, Padova, Italy
| | - Assunta Pozzuoli
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128, Padova, Italy
| | - Carlo Biz
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
| | - Pietro Ruggieri
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology, and Gastroenterology DiSCOG, University-Hospital of Padova, 35128, Padova, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, Padova, 35100, Italy
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, Rutgers, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Marco Sandri
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy
| | - Manuela Basso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Maria Pennuto
- Department of Biomedical Sciences (DBS), University of Padova, 35131, Padova, Italy.
- Veneto Institute of Molecular Medicine (VIMM), Padova, 35100, Italy.
- Padova Neuroscience Center (PNC), Padova, 35100, Italy.
- Dulbecco Telethon Institute (DTI) at the Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy.
| |
Collapse
|
31
|
Zhernov YV, Simanduyev MY, Zaostrovtseva OK, Semeniako EE, Kolykhalova KI, Fadeeva IA, Kashutina MI, Vysochanskaya SO, Belova EV, Shcherbakov DV, Sukhov VA, Sidorova EA, Mitrokhin OV. Molecular Mechanisms of Scombroid Food Poisoning. Int J Mol Sci 2023; 24:ijms24010809. [PMID: 36614252 PMCID: PMC9821622 DOI: 10.3390/ijms24010809] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
Scombroid food poisoning (SFP) is a foodborne disease that develops after consumption of fresh fish and, rarely, seafood that has fine organoleptic characteristics but contains a large amount of exogenous histamine. SFP, like other food pseudo-allergic reactions (FPA), is a disorder that is clinically identical to allergic reactions type I, but there are many differences in their pathogenesis. To date, SFP has been widespread throughout the world and is an urgent problem, although exact epidemiological data on incidence varies greatly. The need to distinguish SFP from true IgE-associated allergy to fish and seafood is one of the most difficult examples of the differential diagnosis of allergic conditions. The most important difference is the absence of an IgE response in SFP. The pathogenesis of SFP includes a complex system of interactions between the body and chemical triggers such as exogenous histamine, other biogenic amines, cis-urocanic acid, salicylates, and other histamine liberators. Because of the wide range of molecular pathways involved in this process, it is critical to understand their differences. This may help predict and prevent poor outcomes in patients and contribute to the development of adequate hygienic rules and regulations for seafood product safety. Despite the vast and lengthy history of research on SFP mechanisms, there are still many blank spots in our understanding of this condition. The goals of this review are to differentiate various molecular mechanisms of SFP and describe methods of hygienic regulation of some biogenic amines that influence the concentration of histamine in the human body and play an important role in the mechanism of SFP.
Collapse
Affiliation(s)
- Yury V. Zhernov
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia
- Center for Medical Anthropology, N.N. Miklukho-Maclay Institute of Ethnology and Anthropology, Russian Academy of Sciences, 119017 Moscow, Russia
- Department of Medical and Biological Disciplines, Reaviz Medical University, 107564 Moscow, Russia
- Correspondence: ; Tel.: +7-(915)-1552000
| | - Mark Y. Simanduyev
- The Baku Branch, I.M. Sechenov First Moscow State Medical University (Sechenov University), Baku AZ1141, Azerbaijan
| | - Olga K. Zaostrovtseva
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Ekaterina E. Semeniako
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Kseniia I. Kolykhalova
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Inna A. Fadeeva
- Department of Foreign Language, Faculty of World Economy, Diplomatic Academy of the Russian Foreign Ministry, 119034 Moscow, Russia
- Department of Public Administration in Foreign Policy, Diplomatic Academy of the Russian Foreign Ministry, 119034 Moscow, Russia
| | - Maria I. Kashutina
- Loginov Moscow Clinical Scientific and Practical Center, 111123 Moscow, Russia
- Department of Public Health Promotion, National Research Centre for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Department of Therapy, Clinical Pharmacology and Emergency Medicine, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| | - Sonya O. Vysochanskaya
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Elena V. Belova
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Denis V. Shcherbakov
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Vitaly A. Sukhov
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Ekaterina A. Sidorova
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Oleg V. Mitrokhin
- Department of General Hygiene, F. Erismann Institute of Public Health, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| |
Collapse
|
32
|
Abstract
The skeletal muscle proteome consists of a large number of diverse protein species with a broad and dynamic concentration range. Since mature skeletal muscles are characterized by a distinctive combination of contractile cells with differing physiological and biochemical properties, it is essential to determine specific differences in the protein composition of fast, slow, and hybrid fibers. Fluorescence two-dimensional difference gel electrophoresis (2D-DIGE) is a powerful comparative tool to analyze fiber type-specific differences between predominantly fast contracting versus slower twitching muscles. In this chapter, the application of the 2D-DIGE method for the comparative analysis of different subtypes of skeletal muscles is outlined in detail. A standardized proteomic workflow is described, involving sample preparation, protein extraction, differential fluorescence labeling using a 3-CyDye system, first-dimension isoelectric focusing, second-dimension slab gel electrophoresis, 2D-DIGE image analysis, protein digestion, and mass spectrometry.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland.
| |
Collapse
|
33
|
Forzano I, Mone P, Mottola G, Kansakar U, Salemme L, De Luca A, Tesorio T, Varzideh F, Santulli G. Efficacy of the New Inotropic Agent Istaroxime in Acute Heart Failure. J Clin Med 2022; 11:7503. [PMID: 36556120 PMCID: PMC9786901 DOI: 10.3390/jcm11247503] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Current therapeutic strategies for acute heart failure (AHF) are based on traditional inotropic agents that are often associated with untoward effects; therefore, finding new effective approaches with a safer profile is dramatically needed. Istaroxime is a novel compound, chemically unrelated to cardiac glycosides, that is currently being studied for the treatment of AHF. Its effects are essentially related to its inotropic and lusitropic positive properties exerted through a dual mechanism of action: activation of the sarcoplasmic reticulum Ca2+ ATPase isoform 2a (SERCA2a) and inhibition of the Na+/K+-ATPase (NKA) activity. The advantages of istaroxime over the available inotropic agents include its lower arrhythmogenic action combined with its capability of increasing systolic blood pressure without augmenting heart rate. However, it has a limited half-life (1 hour) and is associated with adverse effects including pain at the injection site and gastrointestinal issues. Herein, we describe the main mechanism of action of istaroxime and we present a systematic overview of both clinical and preclinical trials testing this drug, underlining the latest insights regarding its adoption in clinical practice for AHF.
Collapse
Affiliation(s)
- Imma Forzano
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Pasquale Mone
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Gaetano Mottola
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Urna Kansakar
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Luigi Salemme
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, University of Campania “Vanvitelli”, 81100 Caserta, Italy
| | - Tullio Tesorio
- Casa di Cura “Montevergine”, Mercogliano, 83013 Avellino, Italy
| | - Fahimeh Varzideh
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
| | - Gaetano Santulli
- Division of Cardiology, Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- Division of Cardiology, Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY 10461, USA
- Department of Molecular Pharmacology, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
34
|
Functional Insights into Protein Kinase A (PKA) Signaling from C. elegans. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111878. [PMID: 36431013 PMCID: PMC9692727 DOI: 10.3390/life12111878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Protein kinase A (PKA), which regulates a diverse set of biological functions downstream of cyclic AMP (cAMP), is a tetramer consisting of two catalytic subunits (PKA-C) and two regulatory subunits (PKA-R). When cAMP binds the PKA-R subunits, the PKA-C subunits are released and interact with downstream effectors. In Caenorhabditis elegans (C. elegans), PKA-C and PKA-R are encoded by kin-1 and kin-2, respectively. This review focuses on the contributions of work in C. elegans to our understanding of the many roles of PKA, including contractility and oocyte maturation in the reproductive system, lipid metabolism, physiology, mitochondrial function and lifespan, and a wide variety of behaviors. C. elegans provides a powerful genetic platform for understanding how this kinase can regulate an astounding variety of physiological responses.
Collapse
|
35
|
Heemstra LA, Koch LG, Britton SL, Novak CM. Altered skeletal muscle sarco-endoplasmic reticulum Ca 2+-ATPase calcium transport efficiency after a thermogenic stimulus. Am J Physiol Regul Integr Comp Physiol 2022; 323:R628-R637. [PMID: 36094445 PMCID: PMC9602703 DOI: 10.1152/ajpregu.00173.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 01/22/2023]
Abstract
Exposure to predator threat induces a rapid and robust increase in skeletal muscle thermogenesis in rats. The central nervous system relays threat information to skeletal muscle through activation of the sympathetic nervous system, but muscle mechanisms mediating this thermogenesis remain unidentified. Given the relevance of sarcolipin-mediated futile calcium cycling through the sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) pump to mammalian muscle nonshivering thermogenesis, we hypothesized that this plays a role in contextually induced muscle thermogenesis as well. This was assessed by measuring enzymatic activity of SERCA and sarcoplasmic reticulum Ca2+ transport, where the apparent coupling ratio (Ca2+ uptake rate divided by ATPase activity rate at a standard Ca2+ concentration) was predicted to decrease in association with muscle thermogenesis. Sprague-Dawley rats exposed to predator (ferret) odor (PO) showed a rapid decrease in the apparent coupling ratio in the soleus muscle, indicating SERCA uncoupling compared with control-odor-exposed rats. A rat model of high aerobic fitness and elevated muscle thermogenesis also demonstrated soleus muscle SERCA uncoupling relative to their obesity-prone, low-fitness counterparts. Both the high- and low-aerobic fitness rats showed soleus SERCA uncoupling with exposure to PO. Finally, no increase in sarcolipin expression in soleus muscle was detected with PO exposure. This dataset implicates muscle uncoupling of SERCA Ca2+ transport and ATP hydrolysis, likely through altered SERCA or sarcolipin function outside of translational regulation, as one contributor to the muscle thermogenesis provoked by exposure to predator threat. These data support the involvement of SERCA uncoupling in both muscle thermogenic induction and enhanced aerobic capacity.
Collapse
Affiliation(s)
- Lydia A Heemstra
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Lauren G Koch
- Department of Physiology and Pharmacology, The University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Steven L Britton
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Colleen M Novak
- Department of Biological Sciences, Kent State University, Kent, Ohio
- School of Biomedical Sciences, Kent State University, Kent, Ohio
| |
Collapse
|
36
|
Enomoto A, Fukasawa T. The role of calcium-calpain pathway in hyperthermia. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:1005258. [PMID: 39086981 PMCID: PMC11285567 DOI: 10.3389/fmmed.2022.1005258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/12/2022] [Indexed: 08/02/2024]
Abstract
Hyperthermia is a promising anticancer treatment modality. Heat stress stimulates proteolytic machineries to regulate cellular homeostasis. Calpain, an intracellular calcium (Ca2+)-dependent cysteine protease, is a modulator that governs various cellular functions. Hyperthermia induces an increase in cytosolic Ca2+ levels and triggers calpain activation. Contrastingly, pre-exposure of cells to mild hyperthermia induces thermotolerance due to the presence of cellular homeostatic processes such as heat shock response and autophagy. Recent studies suggest that calpain is a potential key molecule that links autophagy and apoptosis. In this review, we briefly introduce the regulation of intracellular Ca2+ homeostasis, basic features of calpains with their implications in cancer, immune responses, and the roles and cross-talk of calpains in cellular protection and cell death in hyperthermia.
Collapse
Affiliation(s)
- Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takemichi Fukasawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
37
|
Cho JS, Han YS, Jensen C, Sieck G. Effects of arginase inhibition on myocardial Ca 2+ and contractile responses. Physiol Rep 2022; 10:e15396. [PMID: 35866269 PMCID: PMC9305075 DOI: 10.14814/phy2.15396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 04/18/2023] Open
Abstract
Nitric oxide (NO) is thought to increase cardiac contractility by increasing cytosolic Ca2+ concentration ([Ca2+ ]cyt ) during excitation. Alternatively, NO could increase the sensitivity of the contractile response to [Ca2+ ]cyt (Ca2+ sensitivity). Arginase regulates NO production by competing with NO synthase (NOS), and thus, arginase inhibition should increase cardiac contractility by increasing NO production. We hypothesized that arginase inhibition increases cardiac contractility by increasing both [Ca2+ ]cyt and Ca2+ sensitivity. [Ca2+ ]cyt and contractile (sarcomere length [SL] shortening) responses to electrical stimulation were measured simultaneously in isolated rat cardiomyocytes using an IonOptix system. In the same cardiomyocytes, measurements were obtained at baseline, following 3-min exposure to an arginase inhibitor (S-[2-boronoethyl]-l-cysteine; BEC) and following 3-min exposure to BEC plus a NOS inhibitor (NG -nitro-l-arginine-methyl ester; l-NAME). These responses were compared to time-matched control cardiomyocytes that were untreated. Compared to baseline, BEC increased the amplitude and the total amount of evoked [Ca2+ ]cyt , and the extent and velocity of SL shortening in cardiomyocytes, whereas addition of l-NAME mitigated these effects. The [Ca2+ ]cyt at 50% contraction and relaxation were not different across treatment groups indicating no effect of BEC on Ca2+ sensitivity. The [Ca2+ ]cyt and SL shortening responses in time-matched controls did not vary with time. Arginase inhibition by BEC significantly increased the amplitude and the total amount of evoked [Ca2+ ]cyt , and the extent and velocity of SL shortening in cardiomyocytes, but did not affect Ca2+ sensitivity. These effects of BEC were mitigated by l-NAME. Together, these results indicate an effect of NO on [Ca2+ ]cyt responses that then increase the contractile response of cardiomyocytes.
Collapse
Affiliation(s)
- Jin Sun Cho
- Department of Anesthesiology and Pain MedicineYonsei University College of MedicineSeoulRepublic of Korea
| | - Young Soo Han
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Cole Jensen
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| | - Gary Sieck
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
38
|
Mengeste AM, Katare P, Dalmao Fernandez A, Lund J, Bakke HG, Baker D, Bartesaghi S, Peng XR, Rustan AC, Thoresen GH, Kase ET. Knockdown of sarcolipin (SLN) impairs substrate utilization in human skeletal muscle cells. Mol Biol Rep 2022; 49:6005-6017. [PMID: 35364719 PMCID: PMC9270280 DOI: 10.1007/s11033-022-07387-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/16/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Recent studies have highlighted that uncoupling of sarco-/endoplasmic reticulum Ca2+-ATPase (SERCA) by sarcolipin (SLN) increases ATP consumption and contributes to heat liberation. Exploiting this thermogenic mechanism in skeletal muscle may provide an attractive strategy to counteract obesity and associated metabolic disorders. In the present study, we have investigated the role of SLN on substrate metabolism in human skeletal muscle cells. METHODS AND RESULTS After generation of skeletal muscle cells with stable SLN knockdown (SLN-KD), cell viability, glucose and oleic acid (OA) metabolism, mitochondrial function, as well as gene expressions were determined. Depletion of SLN did not influence cell viability. However, glucose and OA oxidation were diminished in SLN-KD cells compared to control myotubes. Basal respiration measured by respirometry was also observed to be reduced in cells with SLN-KD. The metabolic perturbation in SLN-KD cells was reflected by reduced gene expression levels of peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) and forkhead box O1 (FOXO1). Furthermore, accumulation of OA was increased in cells with SLN-KD compared to control cells. These effects were accompanied by increased lipid formation and incorporation of OA into complex lipids. Additionally, formation of complex lipids and free fatty acid from de novo lipogenesis with acetate as substrate was enhanced in SLN-KD cells. Detection of lipid droplets using Oil red O staining also showed increased lipid accumulation in SLN-KD cells. CONCLUSIONS Overall, our study sheds light on the importance of SLN in maintaining metabolic homeostasis in human skeletal muscle. Findings from the current study suggest that therapeutic strategies involving SLN-mediated futile cycling of SERCA might have significant implications in the treatment of obesity and associated metabolic disorders.
Collapse
Affiliation(s)
- Abel M Mengeste
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway.
| | - Parmeshwar Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| | - Andrea Dalmao Fernandez
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| | - Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| | - Hege G Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| | - David Baker
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Stefano Bartesaghi
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316, Oslo, Norway
| |
Collapse
|
39
|
Ma L, Zou R, Shi W, Zhou N, Chen S, Zhou H, Chen X, Wu Y. SGLT2 inhibitor dapagliflozin reduces endothelial dysfunction and microvascular damage during cardiac ischemia/reperfusion injury through normalizing the XO-SERCA2-CaMKII-coffilin pathways. Am J Cancer Res 2022; 12:5034-5050. [PMID: 35836807 PMCID: PMC9274739 DOI: 10.7150/thno.75121] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/13/2022] [Indexed: 01/12/2023] Open
Abstract
Background: Given the importance of microvascular injury in infarct formation and expansion, development of therapeutic strategies for microvascular protection against myocardial ischemia/reperfusion injury (IRI) is of great interest. Here, we explored the molecular mechanisms underlying the protective effects of the SGLT2 inhibitor dapagliflozin (DAPA) against cardiac microvascular dysfunction mediated by IRI. Methods: DAPA effects were evaluated both in vivo, in mice subjected to IRI, and in vitro, in human coronary artery endothelial cells (HCAECs) exposed to hypoxia/reoxygenation (H/R). DAPA pretreatment attenuated luminal stenosis, endothelial swelling, and inflammation in cardiac microvessels of IRI-treated mice. Results: In H/R-challenged HCAECs, DAPA treatment improved endothelial barrier function, endothelial nitric oxide synthase (eNOS) activity, and angiogenic capacity, and inhibited H/R-induced apoptosis by preventing cofilin-dependent F-actin depolymerization and cytoskeletal degradation. Inhibition of H/R-induced xanthine oxidase (XO) activation and upregulation, sarco(endo)plasmic reticulum calcium-ATPase 2 (SERCA2) oxidation and inactivation, and cytoplasmic calcium overload was further observed in DAPA-treated HCAECs. DAPA also suppressed calcium/Calmodulin (CaM)-dependent kinase II (CaMKII) activation and cofilin phosphorylation, and preserved cytoskeleton integrity and endothelial cell viability following H/R. Importantly, the beneficial effects of DAPA on cardiac microvascular integrity and endothelial cell survival were largely prevented in IRI-treated SERCA2-knockout mice. Conclusions: These results indicate that DAPA effectively reduces cardiac microvascular damage and endothelial dysfunction during IRI through inhibition of the XO-SERCA2-CaMKII-cofilin pathway.
Collapse
Affiliation(s)
- Li Ma
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Rongjun Zou
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wanting Shi
- Department of Paediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Na Zhou
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shaoxian Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xinxin Chen
- Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yueheng Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China.,✉ Corresponding authors: Hao Zhou, E-mail: ; Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China. Xinxin Chen, E-mail: ; Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Heart Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China. Yueheng Wu, E-mail: ; Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of South China Structural Heart Disease, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
40
|
Nemirovskaya TL, Sharlo KA. Roles of ATP and SERCA in the Regulation of Calcium Turnover in Unloaded Skeletal Muscles: Current View and Future Directions. Int J Mol Sci 2022; 23:ijms23136937. [PMID: 35805949 PMCID: PMC9267070 DOI: 10.3390/ijms23136937] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
A decrease in skeletal muscle contractile activity or its complete cessation (muscle unloading or disuse) leads to muscle fibers’ atrophy and to alterations in muscle performance. These changes negatively affect the quality of life of people who, for one reason or another, are forced to face a limitation of physical activity. One of the key regulatory events leading to the muscle disuse-induced changes is an impairment of calcium homeostasis, which leads to the excessive accumulation of calcium ions in the sarcoplasm. This review aimed to analyze the triggering mechanisms of calcium homeostasis impairment (including those associated with the accumulation of high-energy phosphates) under various types of muscle unloading. Here we proposed a hypothesis about the regulatory mechanisms of SERCA and IP3 receptors activity during muscle unloading, and about the contribution of these mechanisms to the excessive calcium ion myoplasmic accumulation and gene transcription regulation via excitation–transcription coupling.
Collapse
|
41
|
Paschou M, Papazafiri P, Charalampous C, Zachariadis M, Dedos SG, Doxakis E. Neuronal microRNAs safeguard ER Ca 2+ homeostasis and attenuate the unfolded protein response upon stress. Cell Mol Life Sci 2022; 79:373. [PMID: 35727337 PMCID: PMC11073139 DOI: 10.1007/s00018-022-04398-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/23/2022] [Accepted: 05/21/2022] [Indexed: 11/30/2022]
Abstract
Ca2+ is a critical mediator of neurotransmitter release, synaptic plasticity, and gene expression, but also excitotoxicity. Ca2+ signaling and homeostasis are coordinated by an intricate network of channels, pumps, and calcium-binding proteins, which must be rapidly regulated at all expression levels. Τhe role of neuronal miRNAs in regulating ryanodine receptors (RyRs) and inositol 1,4,5-triphosphate receptors (IP3Rs) was investigated to understand the underlying mechanisms that modulate ER Ca2+ release. RyRs and IP3Rs are critical in mounting and propagating cytosolic Ca2+ signals by functionally linking the ER Ca2+ content, while excessive ER Ca2+ release via these receptors is central to the pathophysiology of a wide range of neurological diseases. Herein, two brain-restricted microRNAs, miR-124-3p and miR-153-3p, were found to bind to RyR1-3 and IP3R3 3'UTRs, and suppress their expression at both the mRNA and protein level. Ca2+ imaging studies revealed that overexpression of these miRNAs reduced ER Ca2+ release upon RyR/IP3R activation, but had no effect on [Ca2+]i under resting conditions. Interestingly, treatments that cause excessive ER Ca2+ release decreased expression of these miRNAs and increased expression of their target ER Ca2+ channels, indicating interdependence of miRNAs, RyRs, and IP3Rs in Ca2+ homeostasis. Furthermore, by maintaining the ER Ca2+ content, miR-124 and miR-153 reduced cytosolic Ca2+ overload and preserved protein-folding capacity by attenuating PERK signaling. Overall, this study shows that miR-124-3p and miR-153-3p fine-tune ER Ca2+ homeostasis and alleviate ER stress responses.
Collapse
Affiliation(s)
- Maria Paschou
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
| | - Panagiota Papazafiri
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
| | - Chrysanthi Charalampous
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece
| | - Michael Zachariadis
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece
- Material and Chemical Characterization Facility (MC2), Faculty of Science, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Skarlatos G Dedos
- Department of Biology, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, 15784, Athens, Greece.
| | - Epaminondas Doxakis
- Center for Basic Research, Biomedical Research Foundation, Academy of Athens (BRFAA), Soranou Efesiou 4, 11527, Athens, Greece.
| |
Collapse
|
42
|
Ovchinnikov AN, Paoli A, Seleznev VV, Deryugina AV. Royal jelly plus coenzyme Q10 supplementation improves high-intensity interval exercise performance via changes in plasmatic and salivary biomarkers of oxidative stress and muscle damage in swimmers: a randomized, double-blind, placebo-controlled pilot trial. J Int Soc Sports Nutr 2022; 19:239-257. [PMID: 35813842 PMCID: PMC9261740 DOI: 10.1080/15502783.2022.2086015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Background Excessive production of free radicals caused by many types of exercise results in oxidative stress, which leads to muscle damage, fatigue, and impaired performance. Supplementation with royal jelly (RJ) or coenzyme Q10 (CoQ10) has been shown to attenuate exercise-induced oxidant stress in damaged muscle and improve various aspects of exercise performance in many but not all studies. Nevertheless, the effects of treatments based on RJ plus CoQ10 supplementation, which may be potentially beneficial for reducing oxidative stress and enhancing athletic performance, remain unexplored. This study aimed to examine whether oral RJ and CoQ10 co-supplementation could improve high-intensity interval exercise (HIIE) performance in swimmers, inhibiting exercise-induced oxidative stress and muscle damage. Methods Twenty high-level swimmers were randomly allocated to receive either 400 mg of RJ and 60 mg of CoQ10 (RJQ) or matching placebo (PLA) once daily for 10 days. Exercise performance was evaluated at baseline, and then reassessed at day 10 of intervention, using a HIIE protocol. Diene conjugates (DC), Schiff bases (SB), and creatine kinase (CK) were also measured in blood plasma and saliva before and immediately after HIIE in both groups. Results HIIE performance expressed as number of points according to a single assessment system developed and approved by the International Swimming Federation (FINA points) significantly improved in RJQ group (p = 0.013) compared to PLA group. Exercise-induced increase in DC, SB, and CK levels in plasma and saliva significantly diminished only in RJQ group (p < 0.05). Regression analysis showed that oral RJQ administration for 10 days was significantly associated with reductions in HIIE-induced increases in plasmatic and salivary DC, SB, and CK levels compared to PLA. Principal component analysis revealed that swimmers treated with RJQ are grouped by both plasmatic and salivary principal components (PC) into a separate cluster compared to PLA. Strong negative correlation between the number of FINA points and plasmatic and salivary PC1 values was observed in both intervention groups. Conclusion The improvements in swimmers’ HIIE performance were due in significant part to RJQ-induced reducing in lipid peroxidation and muscle damage in response to exercise. These findings suggest that RJQ supplementation for 10 days is potentially effective for enhancing HIIE performance and alleviating oxidant stress. Abbreviations RJ, royal jelly; CoQ10, coenzyme Q10; HIIE, high-intensity interval exercise; DC, diene conjugates; SB, Schiff bases; CK, creatine kinase; RJQ, royal jelly plus coenzyme Q10; PLA, placebo; FINA points, points according to a single assessment system developed and approved by the International Swimming Federation; ROS, reactive oxygen species; 10H2DA, 10-hydroxy-2-decenoic acid; AMPK, 5′-AMP-activated protein kinase; FoxO3, forkhead box O3; MnSOD, manganese-superoxide dismutase; CAT, catalase; E, optical densities; PCA, principal component analysis; PC, principal component; MCFAs, medium-chain fatty acids; CaMKKβ, Ca2+/calmodulin-dependent protein kinase β; TBARS, thiobarbituric acid reactive substances; MDA, malondialdehyde.
Collapse
Affiliation(s)
- Aleksandr N. Ovchinnikov
- Department of Sports Medicine and Psychology, Lobachevsky University, Nizhny Novgorod, Russia
- Laboratory of Integral Human Health, Lobachevsky University, Nizhny Novgorod, Russia
| | - Antonio Paoli
- Laboratory of Integral Human Health, Lobachevsky University, Nizhny Novgorod, Russia
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Vladislav V. Seleznev
- Department of Theory and Methodology of Sport Training, Lobachevsky University, Nizhny Novgorod, Russia
| | - Anna V. Deryugina
- Laboratory of Integral Human Health, Lobachevsky University, Nizhny Novgorod, Russia
- Department of Physiology and Anatomy, Lobachevsky University, Nizhny Novgorod, Russia
| |
Collapse
|
43
|
Sood A, Singh G, Singh TG, Gupta K. Pathological role of the calcium-sensing receptor in sepsis-induced hypotensive shock: Therapeutic possibilities and unanswered questions. Drug Dev Res 2022; 83:1241-1245. [PMID: 35689439 DOI: 10.1002/ddr.21959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 11/09/2022]
Abstract
Sepsis is a life-threatening disease involving multiorgan dysfunction, prompted by an unregulated host response to infection. Shock is a complication of sepsis in which the circulatory and cellular metabolism anomalies are significant enough to raise the risk of death. Calcium dyshomeostasis occurs during sepsis condition due to imbalance between calcium uptake and excessive release induced by inflammatory cytokines. This calcium imbalance can cause activation of calcium-sensing receptors (CaSRs) located on the surface of T cells and thereby promote release of reactive oxygen species (ROS). The elevated ROS and inflammatory cytokines during sepsis condition have been reported to directly damage the endothelial cells, disrupt the barrier functions that might result in leakage of fluids, and inflammatory cells in tissues Moreover, several evidence have revealed that the calcium mediated activation of CaSR could produce systemic vasodilatory response by stimulating the nitric oxide production and opening of calcium-activated potassium channels, while infusion of its antagonist elevated the blood pressure. These evidence indicate that activation of CaSR during sepsis conditions results in release of ROS and inflammatory cytokines, which could produce an endothelial barrier damage, cardiomyocyte apoptosis. These pathological events could produce loss of fluid in tissues and cardiac dysfunction. Further the direct vasodilatory effects of CaSR activation might add to the shock-like condition. Thus, we hereby propose that inhibition of CaSR could suppress the release of ROS, inflammatory mediators, and thereby prevent the endothelial damage, cardiac dysfunction, and maintain systemic vascular tone.
Collapse
Affiliation(s)
- Ankita Sood
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Gaaminepreet Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Thakur G Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Kirti Gupta
- Department of Pharmacology, Maharishi Markandeshwar University, Mullana, Ambala, Haryana, India
| |
Collapse
|
44
|
Understanding the Role of SERCA2a Microdomain Remodeling in Heart Failure Induced by Obesity and Type 2 Diabetes. J Cardiovasc Dev Dis 2022; 9:jcdd9050163. [PMID: 35621874 PMCID: PMC9147026 DOI: 10.3390/jcdd9050163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/29/2022] Open
Abstract
Obesity and type 2 diabetes (T2D) are on trend to become a huge burden across all ages. They cause harm to almost every organ, especially the heart. For decades, the incidence of heart failure with impaired diastolic function (or called heart failure with preserved ejection fraction, HFpEF) has increased sharply. More and more studies have uncovered obesity and T2D to be closely associated with HFpEF. The sarcoplasmic/endoplasmic reticulum calcium ATPase2a (SERCA2a) microdomain is a key regulator of calcium reuptake into the sarcoplasmic reticulum (SR) during diastole. 3′,5′-cyclic adenosine monophosphate (cAMP) and its downstream effector cAMP dependent protein kinase (PKA) act locally within the SERCA2a microdomain to regulate the phosphorylation state of the small regulatory protein phospholamban (PLN), which forms a complex with SERCA2a. When phosphorylated, PLN promotes calcium reuptake into the SR and diastolic cardiac relaxation by disinhibiting SERCA2a pump function. In this review, we will discuss previous studies investigating the PLN/SERCA2a microdomain in obesity and T2D in order to gain a greater understanding of the underlying mechanisms behind obesity- and T2D-induced diastolic dysfunction, with the aim to identify the current state of knowledge and future work that is needed to guide further research in the field.
Collapse
|
45
|
Guarnieri AR, Benson TW, Tranter M. Calcium cycling as a mediator of thermogenic metabolism in adipose tissue. Mol Pharmacol 2022; 102:MOLPHARM-MR-2021-000465. [PMID: 35504660 PMCID: PMC9341262 DOI: 10.1124/molpharm.121.000465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 11/22/2022] Open
Abstract
Canonical non-shivering thermogenesis (NST) in brown and beige fat relies on uncoupling protein 1 (UCP1)-mediated heat generation, although alternative mechanisms of NST have been identified, including sarcoplasmic reticulum (SR)-calcium cycling. Intracellular calcium is a crucial cell signaling molecule for which compartmentalization is tightly regulated, and the sarco-endoplasmic calcium ATPase (SERCA) actively pumps calcium from the cytosol into the SR. In this review, we discuss the capacity of SERCA-mediated calcium cycling as a significant mediator of thermogenesis in both brown and beige adipocytes. Here, we suggest two primary mechanisms of SR calcium mediated thermogenesis. The first mechanism is through direct uncoupling of the ATPase and calcium pump activity of SERCA, resulting in the energy of ATP catalysis being expended as heat in the absence of calcium transport. Regulins, a class of SR membrane proteins, act to decrease the calcium affinity of SERCA and uncouple the calcium transport function from ATPase activity, but remain largely unexplored in adipose tissue thermogenesis. A second mechanism is through futile cycling of SR calcium whereby SERCA-mediated SR calcium influx is equally offset by SR calcium efflux, resulting in ATP consumption without a net change in calcium compartmentalization. A fuller understanding of the functional and mechanistic role of calcium cycling as a mediator of adipose tissue thermogenesis and how manipulation of these pathways can be harnessed for therapeutic gain remains unexplored. Significance Statement Enhancing thermogenic metabolism in brown or beige adipose tissue may be of broad therapeutic utility to reduce obesity and metabolic syndrome. Canonical BAT-mediated thermogenesis occurs via uncoupling protein 1 (UCP1). However, UCP1-independent pathways of thermogenesis, such as sarcoplasmic (SR) calcium cycling, have also been identified, but the regulatory mechanisms and functional significance of these pathways remain largely unexplored. Thus, this mini-review discusses the state of the field with regard to calcium cycling as a thermogenic mediator in adipose tissue.
Collapse
Affiliation(s)
| | - Tyler W Benson
- University of Cincinnati College of Medicine, United States
| | | |
Collapse
|
46
|
Houthaeve G, De Smedt SC, Braeckmans K, De Vos WH. The cellular response to plasma membrane disruption for nanomaterial delivery. NANO CONVERGENCE 2022; 9:6. [PMID: 35103909 PMCID: PMC8807741 DOI: 10.1186/s40580-022-00298-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Delivery of nanomaterials into cells is of interest for fundamental cell biological research as well as for therapeutic and diagnostic purposes. One way of doing so is by physically disrupting the plasma membrane (PM). Several methods that exploit electrical, mechanical or optical cues have been conceived to temporarily disrupt the PM for intracellular delivery, with variable effects on cell viability. However, apart from acute cytotoxicity, subtler effects on cell physiology may occur as well. Their nature and timing vary with the severity of the insult and the efficiency of repair, but some may provoke permanent phenotypic alterations. With the growing palette of nanoscale delivery methods and applications, comes a need for an in-depth understanding of this cellular response. In this review, we summarize current knowledge about the chronology of cellular events that take place upon PM injury inflicted by different delivery methods. We also elaborate on their significance for cell homeostasis and cell fate. Based on the crucial nodes that govern cell fitness and functionality, we give directions for fine-tuning nano-delivery conditions.
Collapse
Affiliation(s)
- Gaëlle Houthaeve
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Ghent University, Ghent, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
47
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
48
|
Nusier M, Shah AK, Dhalla NS. Structure-Function Relationships and Modifications of Cardiac Sarcoplasmic Reticulum Ca2+-Transport. Physiol Res 2022; 70:S443-S470. [DOI: 10.33549/physiolres.934805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Sarcoplasmic reticulum (SR) is a specialized tubular network, which not only maintains the intracellular concentration of Ca2+ at a low level but is also known to release and accumulate Ca2+ for the occurrence of cardiac contraction and relaxation, respectively. This subcellular organelle is composed of several phospholipids and different Ca2+-cycling, Ca2+-binding and regulatory proteins, which work in a coordinated manner to determine its function in cardiomyocytes. Some of the major proteins in the cardiac SR membrane include Ca2+-pump ATPase (SERCA2), Ca2+-release protein (ryanodine receptor), calsequestrin (Ca2+-binding protein) and phospholamban (regulatory protein). The phosphorylation of SR Ca2+-cycling proteins by protein kinase A or Ca2+-calmodulin kinase (directly or indirectly) has been demonstrated to augment SR Ca2+-release and Ca2+-uptake activities and promote cardiac contraction and relaxation functions. The activation of phospholipases and proteases as well as changes in different gene expressions under different pathological conditions have been shown to alter the SR composition and produce Ca2+-handling abnormalities in cardiomyocytes for the development of cardiac dysfunction. The post-translational modifications of SR Ca2+ cycling proteins by processes such as oxidation, nitrosylation, glycosylation, lipidation, acetylation, sumoylation, and O GlcNacylation have also been reported to affect the SR Ca2+ release and uptake activities as well as cardiac contractile activity. The SR function in the heart is also influenced in association with changes in cardiac performance by several hormones including thyroid hormones and adiponectin as well as by exercise-training. On the basis of such observations, it is suggested that both Ca2+-cycling and regulatory proteins in the SR membranes are intimately involved in determining the status of cardiac function and are thus excellent targets for drug development for the treatment of heart disease.
Collapse
Affiliation(s)
| | | | - NS Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen, Research Centre, 351 Tache Avenue, Winnipeg, MB, R2H 2A6 Canada.
| |
Collapse
|
49
|
Kovacs G, Reimer L, Jensen PH. Endoplasmic Reticulum-Based Calcium Dysfunctions in Synucleinopathies. Front Neurol 2021; 12:742625. [PMID: 34744980 PMCID: PMC8563702 DOI: 10.3389/fneur.2021.742625] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022] Open
Abstract
Neuronal calcium dyshomeostasis has been associated to Parkinson's disease (PD) development based on epidemiological studies on users of calcium channel antagonists and clinical trials are currently conducted exploring the hypothesis of increased calcium influx into neuronal cytosol as basic premise. We reported in 2018 an opposite hypothesis based on the demonstration that α-synuclein aggregates stimulate the endoplasmic reticulum (ER) calcium pump SERCA and demonstrated in cell models the existence of an α-synuclein-aggregate dependent neuronal state wherein cytosolic calcium is decreased due to an increased pumping of calcium into the ER. Inhibiting the SERCA pump protected both neurons and an α-synuclein transgenic C. elegans model. This models two cellular states that could contribute to development of PD. First the prolonged state with reduced cytosolic calcium that could deregulate multiple signaling pathways. Second the disease ER state with increased calcium concentration. We will discuss our hypothesis in the light of recent papers. First, a mechanistic study describing how variation in the Inositol-1,4,5-triphosphate (IP3) kinase B (ITPKB) may explain GWAS studies identifying the ITPKB gene as a protective factor toward PD. Here it was demonstrated that how increased ITPKB activity reduces influx of ER calcium to mitochondria via contact between IP3-receptors and the mitochondrial calcium uniporter complex in ER-mitochondria contact, known as mitochondria-associated membranes (MAMs). Secondly, it was demonstrated that astrocytes derived from PD patients contain α-synuclein accumulations. A recent study has demonstrated how human astrocytes derived from a few PD patients carrying the LRRK2-2019S mutation express more α-synuclein than control astrocytes, release more calcium from ER upon ryanodine receptor (RyR) stimulation, show changes in ER calcium channels and exhibit a decreased maximal and spare respiration indicating altered mitochondrial function in PD astrocytes. Here, we summarize the previous findings focusing the effect of α-synuclein to SERCA, RyR, IP3R, MCU subunits and other MAM-related channels. We also consider how the SOCE-related events could contribute to the development of PD.
Collapse
Affiliation(s)
- Gergo Kovacs
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lasse Reimer
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Poul Henning Jensen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
50
|
Sordi G, Goti A, Young HS, Palchetti I, Tadini‐Buoninsegni F. Stimulation of Ca 2+ -ATPase Transport Activity by a Small-Molecule Drug. ChemMedChem 2021; 16:3293-3299. [PMID: 34297466 PMCID: PMC8571031 DOI: 10.1002/cmdc.202100350] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Indexed: 11/11/2022]
Abstract
The sarco(endo)plasmic reticulum Ca2+ -ATPase (SERCA) hydrolyzes ATP to transport Ca2+ from the cytoplasm to the sarcoplasmic reticulum (SR) lumen, thereby inducing muscle relaxation. Dysfunctional SERCA has been related to various diseases. The identification of small-molecule drugs that can activate SERCA may offer a therapeutic approach to treat pathologies connected with SERCA malfunction. Herein, we propose a method to study the mechanism of interaction between SERCA and novel SERCA activators, i. e. CDN1163, using a solid supported membrane (SSM) biosensing approach. Native SR vesicles or reconstituted proteoliposomes containing SERCA were adsorbed on the SSM and activated by ATP concentration jumps. We observed that CDN1163 reversibly interacts with SERCA and enhances ATP-dependent Ca2+ translocation. The concentration dependence of the CDN1163 effect provided an EC50 =6.0±0.3 μM. CDN1163 was shown to act directly on SERCA and to exert its stimulatory effect under physiological Ca2+ concentrations. These results suggest that CDN1163 interaction with SERCA can promote a protein conformational state that favors Ca2+ release into the SR lumen.
Collapse
Affiliation(s)
- Giacomo Sordi
- Department of Chemistry “Ugo Schiff”University of FlorenceVia della Lastruccia 3–1350019Sesto FiorentinoItaly
- Present address: PQE Group50066 ReggelloFlorenceItaly
| | - Andrea Goti
- Department of Chemistry “Ugo Schiff”University of FlorenceVia della Lastruccia 3–1350019Sesto FiorentinoItaly
| | - Howard S. Young
- Department of BiochemistryUniversity of AlbertaEdmonton, AlbertaT6G 2H7Canada
| | - Ilaria Palchetti
- Department of Chemistry “Ugo Schiff”University of FlorenceVia della Lastruccia 3–1350019Sesto FiorentinoItaly
| | | |
Collapse
|